1
|
Wu B, Yao C, Wang H, Dai H, Tian B, Li D, Xu J, Cheng H, Xu F, Sun D, Wang C. Ellagic acid-protein nano-complex inhibits tumor growth by reducing the intratumor bacteria and inhibiting histamine production. Biomaterials 2025; 317:123078. [PMID: 39753083 DOI: 10.1016/j.biomaterials.2024.123078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/25/2024] [Accepted: 12/30/2024] [Indexed: 02/04/2025]
Abstract
In recent years, there has been growing interest in understanding the role of bacteria within tumors and their potential as targets for cancer therapy. In this work, we developed an ellagic acid (EA) - endogenous protein (eP) nanocomposite (eP-EA) to target tumors by EPR (enhanced permeability and retention), kill bacteria within tumors to regulate anti-tumor immune responses. The potential mechanism of eP-EA treatment is associated with the reduced abundance and diversity of microorganisms within the tumor, culminating with an altered metabolism within the Tumor microenvironment (TME). Among them, the metabolite histamine that contributes to tumor progression, is significantly reduced in the TME after eP-EA treatment. We show that one possible mechanism by which these microbes promote tumor growth is through the production of histamine. This work suggests that the ellagic acid (EA)-protein nano complex can enhance cancer immunotherapy by targeting the intratumoral bacteria and reduce their production of histamine, delineating the potential relationship between intratumor bacteria and their impact on tumors. Our work suggests that the EA-protein nano complex can enhance cancer immunotherapy by targeting the intratumoral bacteria, suggesting the role of bacterial metabolites in contributing to tumor progression.
Collapse
Affiliation(s)
- Bingbing Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Chenlu Yao
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Heng Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Huaxing Dai
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Bo Tian
- The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, China
| | - Dongxiao Li
- The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, China
| | - Jialu Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Haibo Cheng
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Fang Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Dongdong Sun
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
2
|
Li Z, Li J, Bai X, Huang X, Wang Q. Tumor microenvironment as a complex milieu driving cancer progression: a mini review. Clin Transl Oncol 2025; 27:1943-1952. [PMID: 39342061 PMCID: PMC12033186 DOI: 10.1007/s12094-024-03697-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 08/22/2024] [Indexed: 10/01/2024]
Abstract
It has been spotlighted that the Tumor Microenvironment (TME) is crucial for comprehending cancer progression and therapeutic resistance. Therefore, this comprehensive review elucidates the intricate architecture of the TME, which encompasses tumor cells, immune components, support cells, and a myriad of bioactive molecules. These constituents collectively foster dynamic interactions that underpin tumor growth, metastasis, and nuanced responses to anticancer therapies. Notably, the TME's role extends beyond mere physical support, serving as a critical mediator in cancer-cell evolution, immune modulation, and treatment outcomes. Innovations targeting the TME, including strategies focused on the vasculature, immune checkpoints, and T-cell therapies, have forged new pathways for clinical intervention. However, the heterogeneity and complexity of the TME present significant challenges, necessitating deeper exploration of its components and their interplay to enhance therapeutic efficacy. This review underscores the imperative for integrated research strategies that amalgamate insights from tumor biology, immunology, and systems biology. Such an approach aims to refine cancer treatments and improve patient prognoses by exploiting the TME's complexity.
Collapse
Affiliation(s)
- Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China.
- National Center for Stomatology, Shanghai, China.
- National Clinical Research Center for Oral Diseases, Shanghai, China.
- Shanghai Key Laboratory of Stomatology, Shanghai, China.
- Shanghai Research Institute of Stomatology, Shanghai, China.
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai, China.
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China.
| | - Jing Li
- Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
| | - Xiaolei Bai
- Department of Stomatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Xufeng Huang
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary.
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China.
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Digestive Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
3
|
Ye Z, Gao L, Guo Z, Wang Q. Oral and intestinal flora translocation and tumor development. J Cancer Res Ther 2025; 21:323-333. [PMID: 40317136 DOI: 10.4103/jcrt.jcrt_50_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 02/07/2025] [Indexed: 05/07/2025]
Abstract
ABSTRACT Cancer metastasis is the leading cause of death in patients. In recent years, there has been a growing recognition of the role of tumor-associated microflora in tumor metastasis. The connection between oral and gut microflora and the tumor microenvironment has also been extensively studied. The migration of oral and gut microflora is closely associated with tumor development. Although there is awareness regarding the significant impact of microbial communities on human health, the focus on their relationship with host organisms, particularly those related to tumor-associated microflora, remains inadequate. As an integral part of the body, the host microflora is crucial for regulating the cancer risk and preventing tumor recurrence. The oral-gut axis plays an indispensable role in human immunity, and many types of cancers, such as colorectal, pancreatic, and breast, are significantly influenced by their internal microbial communities. However, further exploration into the mechanisms underlying the role of the intratumoral microflora in cancer is necessary to achieve a comprehensive understanding. We have summarized and analyzed related articles in PubMed. This article reviews the impact of the oral-gut axis on the human immune system, explores the relationship between the translocation of the oral and intestinal flora and the tumor microenvironment, analyzes the specific mechanisms involved in the translocation of the oral and intestinal microflora during the evolution and progression of tumors, and elaborates on the correlations between the occurrence and development of tumors and the changes in the microflora. Finally, a summary of these abovementioned points is provided.
Collapse
Affiliation(s)
- Zhiyuan Ye
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Linglin Gao
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Zhi Guo
- Department of Hematology, The 6 Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Qiang Wang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Kan L, Yu Y, Wang Y, Shi L, Fan T, Chen H, Ren C. The application of organoids in investigating immune evasion in the microenvironment of gastric cancer and screening novel drug candidates. Mol Cancer 2025; 24:125. [PMID: 40287758 PMCID: PMC12032790 DOI: 10.1186/s12943-025-02328-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Gastric cancer (GC) is a prevalent digestive system tumor, the fifth most diagnosed cancer worldwide, and a leading cause of cancer deaths. GC is distinguished by its pronounced heterogeneity and a dynamically evolving tumor microenvironment (TME). The lack of accurate disease models complicates the understanding of its mechanisms and impedes the discovery of novel drugs. A growing body of evidence suggests that GC organoids, developed using organoid culture technology, preserve the genetic, phenotypic, and behavioral characteristics. GC organoids hold significant potential for predicting treatment responses in individual patients. This review provides a comprehensive overview of the current clinical treatment strategies for GC, as well as the history, construction and clinical applications of organoids. The focus is on the role of organoids in simulating the TME to explore mechanisms of immune evasion and intratumoral microbiota in GC, as well as their applications in guiding clinical drug therapy and facilitating novel drug screening. Furthermore, we summarize the limitations of GC organoid models and underscore the need for continued technological advancements to benefit both basic and translational oncological research.
Collapse
Affiliation(s)
- Liuyue Kan
- Department of Laboratory Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Ying Yu
- Department of Laboratory Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Yaxue Wang
- Department of Laboratory Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Lei Shi
- Department of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, No. 98 Western Nantong Road, Yangzhou, 225001, China
| | - Tingyuan Fan
- Department of Laboratory Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Hui Chen
- Department of Geriatrics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, No. 98 Western Nantong Road, Yangzhou, 225001, China.
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, No. 98, Western Nantong Road, Yangzhou, 225001, China.
| | - Chuanli Ren
- Department of Laboratory Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China.
- Department of Laboratory Medicine, The Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, China.
- The Yangzhou Clinical Medical College of Xuzhou Medical University, No. 98, Western Nantong Road, Yangzhou, 225001, China.
| |
Collapse
|
5
|
Li YR, Shen X, Zhu Y, Lyu Z, Yang L. The microbiota shapes the life trajectory of mucosal-associated invariant T cells. Trends Microbiol 2025:S0966-842X(25)00107-6. [PMID: 40280795 DOI: 10.1016/j.tim.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025]
Abstract
Mucosal-associated invariant T (MAIT) cells are innate-like T cells predominantly located in barrier tissues such as the lung, liver, skin, and colon. These cells recognize metabolites derived from the riboflavin biosynthetic pathway, which can rapidly traverse epithelial barriers and be presented during MAIT cell differentiation in the thymus and maturation in peripheral tissues. Furthermore, microbial metabolites significantly influence MAIT cell functions in various conditions, including cancer. This review summarizes how the microbiota shapes the life trajectory of MAIT cells and their antitumor reactivity. Additionally, we discuss the therapeutic implications of manipulating the microbiota as a 'bug-drug' strategy to enhance MAIT cell antitumor immunity, particularly in mucosal cancers, while emphasizing challenges and future directions for integrating microbiota considerations into MAIT cell-based therapies.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Xinyuan Shen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yichen Zhu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zibai Lyu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA 90095, USA; Goodman-Luskin Microbiome Center, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
6
|
Liu S, Qi L, Dong L, Sun W, Liu S, Li P, Zhang N. Prognostic implications of the interaction between intratumoral microbiome and immune response in gastric cancer. Microbiol Spectr 2025; 13:e0283024. [PMID: 40202312 PMCID: PMC12054076 DOI: 10.1128/spectrum.02830-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 03/08/2025] [Indexed: 04/10/2025] Open
Abstract
Gastric cancer (GC) prognosis is significantly influenced by intratumoral microbiomes, which modulate host-immune interactions. This study analyzed data from the The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases to identify immune genes associated with GC prognosis and conducted prognostic immune subtypes. GC patients were classified into two distinct prognostic immune phenotypes C1 and C2 based on the non-negative matrix factorization consensus clusters. Phenotype C2 exhibited a better prognosis and distinct immune characteristics, including enhanced presence of Th2 and Th17 cells and improved response to chemotherapy. In contrast, phenotype C1 showed higher expression levels of PDCD1LG2 and TLR9, which were critical immune factors involved in immune regulation. Both phenotypes were linked to immune genes influencing intratumoral microbiomes and GC immunotherapy responses. A prediction risk model was constructed using the LASSO regression analysis and showed great prognostic value for GC patients. The key genes were correlated with immune cells and suppressed the function of the host immune system. The intratumoral microbiomes were strongly associated with the hosts' immune infiltration and significantly interacted with host immune genes to influence GC outcomes. Candidatus Nitrosotenuis plays a significant role in predicting the prognosis of GC patients. This research underscores the pivotal role of intratumoral microbiomes in GC prognosis and supports the development of future personalized therapeutic approaches.IMPORTANCEIncreasing evidence confirms the presence of intratumoral microbiomes. However, the role of the intratumoral microbiomes in the progression of gastric cancer and their relationship with the immune microenvironment remain unclear. Our study classified gastric cancer patients into two immune prognostic subtypes, C1 and C2, using non-negative matrix factorization consensus clusters. The C2 subtype exhibited a better prognosis and more pronounced immune characteristics. Microbiome analyses revealed associations between both subtypes and immune genes that affect intratumoral microbiomes and their responses to immunotherapy. The intratumoral microbiomes were closely linked with host immune infiltration and significantly interacted with immune genes, which influence the prognosis of gastric cancer. Notably, Candidatus Nitrosotenuis showed a significant prognostic value in gastric cancer patients. Our findings highlight the critical role of the intratumoral microbiomes in affecting gastric cancer prognosis and its interaction with the immune microenvironment, supporting future personalized therapeutic approaches.
Collapse
Affiliation(s)
- Sifan Liu
- Department of Gastroenterology, State Key Laboratory for Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesions of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lingyu Qi
- Department of Gastroenterology, State Key Laboratory for Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesions of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lu Dong
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Wenjing Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Siying Liu
- Department of Gastroenterology, State Key Laboratory for Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesions of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Peng Li
- Department of Gastroenterology, State Key Laboratory for Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesions of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Nan Zhang
- Department of Gastroenterology, State Key Laboratory for Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesions of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Zhou C, Zhang Y, Tian B, Yu Y, Li D, Wu B, Chang W, Shi T, Xu F, Bai J, Wang C. Bacteria-responsive cytoderm drug delivery systems. Biomater Sci 2025. [PMID: 40196901 DOI: 10.1039/d5bm00026b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Signs of bacterial activities have been reported in a variety of disease models. Here, we extracted plant cytoderm ghosts (PCGs) from plant cells, acting as bacteria-responsive drug delivery systems (DDSs) that release drugs specifically in response to the presence or activity of bacteria. Cellulose, which is one of the main components of PCGs, can be degraded in the presence of specialized bacteria that secrete enzymes to convert the cellulose into simpler sugars, thus breaking down the structure of PCGs to release the loaded drugs. In our study, PCGs loaded with ciprofloxacin (PCG@CIP) could effectively inhibit the proliferation and retention of bacteria at the infection site, and improve the local wound microenvironment to accelerate wound repair. In addition, the PCG platform with anticancer drugs could effectively regulate the progression of tumor growth. Therefore, we report a new drug delivery system that responds to the microbiota based on plant cytoderm, providing a new option for drug responsive delivery.
Collapse
Affiliation(s)
- Chenmeng Zhou
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Yue Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Bo Tian
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, China.
| | - Yue Yu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Dongxiao Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Bingbing Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Wenju Chang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, China.
| | - Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Fang Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Jinyu Bai
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, China.
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
8
|
Detweiler BMK, Zangirolami AC, Wright GA, Kim DM, Bagnato VS. Enhancement of activity in the Cancer immune system due to the presence of microcomponents when Exposed to Photodynamic: An in Vitro Experiment. Photodiagnosis Photodyn Ther 2025; 52:104518. [PMID: 39929356 DOI: 10.1016/j.pdpdt.2025.104518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/23/2025]
Abstract
The application of photodynamic therapy (PDT) has aimed at destroying tumor cells and microbiological control in infections. Experimentation in vitro with individual elements of a biological system enables the study of mechanism of action and provides directions to in vivo applications where cancer coexists with normal cells and microorganisms, whether opportunistic or not. The action of the combined set of elements leads to a different behavior from each of the components isolated. In this study, we created a simple model to investigate the effects of PDT (1) on the coculture of macrophages (2) and melanoma cells (3) infected with E. coli (4) under different combinations. Comparing the 4 individual elements of the system interacting with each other to all of them interacting together, we found that macrophage phagocytosis depends on the order in which the elements are exposed to PDT. That indicates the behavior of macrophages depends on their relationship with the microenvironment. Applying the photodynamic effect to each component separately or to all of them together results in different outcomes. These in vitro experiments provide pathways to understand or to design new and more efficient applications of PDT.
Collapse
Affiliation(s)
| | | | - Gus A Wright
- Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Da M Kim
- Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Vanderlei S Bagnato
- Biomedical Engineering, Texas A&M University, College Station TX, USA; IFSC- University of São Paulo, São Carlos, SP, Brazil
| |
Collapse
|
9
|
Patra D, Dev G, Hand TW, Overacre-Delgoffe A. Friends close, enemies closer: the complex role of the microbiome in antitumor immunity. Curr Opin Immunol 2025; 93:102537. [PMID: 40015179 DOI: 10.1016/j.coi.2025.102537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 03/01/2025]
Abstract
Immunotherapy has achieved remarkable advances in cancer treatment by harnessing the immune system to combat tumors, yet its effectiveness remains inconsistent across patients and tumor types. The microbiota, a diverse assemblage of microorganisms residing at host barrier surfaces, is pivotal in shaping immune responses. This review explores the direct and indirect mechanisms via which the microbiota modulates antitumor immune responses both locally within the tumor microenvironment and systemically by affecting distant tumors. We discuss recent findings linking microbiota-derived metabolites and microbiota-derived antigens with antitumor immunity and immunotherapy response. Additionally, we discuss recent advances in microbiome-based therapies, including fecal microbiota transplantation. We propose the use and development of new analytical techniques to further characterize the complex functions and interactions between the microbiome and immune system. To conclude, we outline recommendations for future research and therapeutic approaches to leverage the microbiome to improve current immunotherapies.
Collapse
Affiliation(s)
- Dipyaman Patra
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, USA
| | - Gagan Dev
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Timothy W Hand
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA.
| | - Abigail Overacre-Delgoffe
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, USA.
| |
Collapse
|
10
|
Hu MM, Zhao Y, Zhang N, Gong FY, Zhang W, Dong CS, Dai JF, Wang J. Tumor Microenvironment: Obstacles and Opportunities for T Cell-Based Tumor Immunotherapies. Mol Cancer Res 2025; 23:277-287. [PMID: 39898773 DOI: 10.1158/1541-7786.mcr-24-0747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/20/2024] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
The complex composition and dynamic change of the tumor microenvironment (TME), mainly consisting of tumor cells, immune cells, stromal cells, and extracellular components, significantly impede the effector function of cytotoxic T lymphocytes (CTL), thus representing a major obstacle for tumor immunotherapies. In this review, we summarize and discuss the impacts and underlying mechanisms of major elements in the TME (different cell types, extracellular matrix, nutrients and metabolites, etc.) on the infiltration, survival, and effector functions of T cells, mainly CD8+ CTLs. Moreover, we also highlight recent advances that may potentiate endogenous antitumor immunity and improve the efficacy of T cell-based immunotherapies in patients with cancer by manipulating components inside/outside of the TME. A deeper understanding of the effects and action mechanisms of TME components on the tumor-eradicating ability of CTLs may pave the way for discovering new targets to augment endogenous antitumor immunity and for designing combinational therapeutic regimens to enhance the efficacy of tumor immunotherapies in the clinic.
Collapse
Affiliation(s)
- Miao-Miao Hu
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Ying Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Nan Zhang
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Fang-Yuan Gong
- Department of Immunology, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Wei Zhang
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Chun-Sheng Dong
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jian-Feng Dai
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jun Wang
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| |
Collapse
|
11
|
Chang Y, Long M, Shan H, Liu L, Zhong S, Luo JL. Combining gut microbiota modulation and immunotherapy: A promising approach for treating microsatellite stable colorectal cancer. Crit Rev Oncol Hematol 2025; 208:104629. [PMID: 39864533 DOI: 10.1016/j.critrevonc.2025.104629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent and lethal cancers worldwide, ranking third in incidence and second in mortality. While immunotherapy has shown promise in patients with deficient mismatch repair (dMMR) or high microsatellite instability (MSI-H), its effectiveness in proficient mismatch repair (pMMR) or microsatellite stable (MSS) CRC remains limited. Recent advances highlight the gut microbiota as a potential modulator of anti-tumor immunity. The gut microbiome can significantly influence the efficacy of immune checkpoint inhibitors (ICIs), especially in pMMR/MSS CRC, by modulating immune responses and systemic inflammation. This review explores the role of the gut microbiota in pMMR/MSS CRC, the mechanisms by which it may enhance immunotherapy, and current strategies for microbiota modulation. We discuss the potential benefits of combining microbiota-targeting interventions with immunotherapy to improve treatment outcomes for pMMR/MSS CRC patients.
Collapse
Affiliation(s)
- Yujie Chang
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Min Long
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Hanguo Shan
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China
| | - Logen Liu
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China
| | - Shangwei Zhong
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Jun-Li Luo
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, USC, Hunan 410008, China.
| |
Collapse
|
12
|
Chen Y, Wu J, Cai K, Xiao X, Chen Y, Zhang X, Deng S, Pei C, Chen Y, Xie Z, Li P, Liao Q. Bifidobacterium longum subsp. longum XZ01 delays the progression of colon cancer in mice through the interaction between the microbial spatial distribution and tumour immunity. Int Immunopharmacol 2025; 150:114283. [PMID: 39955918 DOI: 10.1016/j.intimp.2025.114283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/25/2025] [Accepted: 02/08/2025] [Indexed: 02/18/2025]
Abstract
Studies have shown that the colonisation of active microorganisms is more conducive to the development of tumour immunotherapy, but intuitive evidence regarding shaping of the tumour immune microenvironment is lacking. In this study, we used Bifidobacterium longum subsp. longum (XZ01) to intervene in a colon cancer mouse model and found that its mechanism may be related to the interaction between the spatial distribution of microorganisms and tumour immunity. Through the visualisation method we established, for the first time, we showed that harmful active bacteria such as Streptococcus and Rhodococcus specifically accumulate in the middle and upper layers of tumour tissue. These bacteria likely participate in signalling pathways that affect macrophages by directly contacting or invading the macrophages, leading to a nondifferentiated state in macrophages and the loss of some immune functions. Furthermore, the accumulation of Streptococcus and Rhodococcus fragments in the deep layer of tumour tissue likely upregulates the expression of IL-10 in tumour tissue and inhibits other immune cells, such as CD8+ T cells, DC and NK cells. In contrast, XZ01 can specifically compete for the growth sites of Streptococcus and Rhodococcus in the middle and upper layers of tumour tissue and probably protects macrophages from being invaded by harmful bacteria. XZ01 directly regulates the polarisation of M0 macrophages towards the M1 phenotype by upregulating IFN-γ, thus activating tumour immunity to inhibit the growth of tumour cells. This study revealed that the influence of active microorganisms on the tumour immune microenvironment is crucial for effective immunotherapy intervention, potentially offering new targets for improving patient prognosis.
Collapse
Affiliation(s)
- Ying Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; School of Pharmaceutical Sciences, Guangdong Yunfu Vocational College of Chinese Medicine, Yunfu 527300, China
| | - Jinyun Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Kaiwei Cai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiaoyi Xiao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Ye Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xingyuan Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Song Deng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Chaoying Pei
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yanlong Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zhiyong Xie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Pei Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Qiongfeng Liao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| |
Collapse
|
13
|
Qi F, Meng K, Zhao X, Lv J, Huang L, Fan X, Feng Z. Targeting gut microbiota: a potential therapeutic approach for tumor microenvironment in glioma. Front Neurol 2025; 16:1549465. [PMID: 40183013 PMCID: PMC11965986 DOI: 10.3389/fneur.2025.1549465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
Glioma, being one of the malignant tumors with the highest mortality rate globally, has an unclear pathogenesis, and the existing treatment effects still have certain limitations. The tumor microenvironment (TME) plays an important role in the occurrence, development, and recurrence of glioma. As one of the important regulatory factors of TME, the gut microbiota can regulate the progression of glioma not only by interacting with the brain through the brain-gut axis but also by influencing the tumor immune microenvironment (TIME) and inflammatory microenvironment. Recent studies have identified the gut microbiota and TME as potential therapeutic targets for glioma. This paper aims to summarize the role of the gut microbiota in TME, the association between them and glioma, and the potential of developing new intervention measures by targeting the gut microbiota. Understanding the involvement process of the gut microbiota in glioma may pave the way for the development of effective treatment methods that can regulate TME and prevent disease progression.
Collapse
Affiliation(s)
- Fan Qi
- College of Integrated Traditional and Western Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Kaiqiang Meng
- College of Traditional Chinese Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Xiaoping Zhao
- Neurosurgery Department of the Encephalopathy Hospital, Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Jing Lv
- College of Integrated Traditional and Western Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Lan Huang
- College of Integrated Traditional and Western Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Xiaoxuan Fan
- College of Integrated Traditional and Western Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Zhaoqun Feng
- Neurosurgery Department of the Encephalopathy Hospital, Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi, China
| |
Collapse
|
14
|
Yang Y, Meng Y, Xu Z, Zhang Q, Li M, Kong F, Zhang S, Li X, Zhu Y. Leveraging microbiome signatures to predict tumor immune microenvironment and prognosis of patients with endometrial carcinoma. Discov Oncol 2025; 16:299. [PMID: 40069468 PMCID: PMC11896907 DOI: 10.1007/s12672-025-02038-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Recent studies suggest that the human microbiome influence tumor development. Endometrial carcinoma (EC) is the sixth most common malignancy in women. Recent research has demonstrated the microbes play a critical role in the development and metastasis of EC. However, it remains unclear whether intratumoral microbes are associated with tumor microenvironment (TME) and prognosis of EC. In this study, we collected the EC microbiome data from cBioPortal and constructed a prognostic model based on Resident Microbiome of Endometrium (RME). We then examined the relationship between the RME score, immune cell infiltration, immunotherapy-related signature, and prognosis. The findings demonstrated the independent prognostic value of the RME score for EC. The group with low RME scores had higher enrichment of immune cells. Drug sensitivity analysis revealed that the RME score may serve as a potential predictor of chemotherapy efficacy. In conclusion, our research offers new perspectives on the relationships between tumor immunity and microbes.
Collapse
Affiliation(s)
- Yuting Yang
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Yuchen Meng
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Ziyang Xu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Qin Zhang
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Miaomiao Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Fanbing Kong
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Suping Zhang
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Xinling Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China.
| | - Yihua Zhu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
15
|
Ma C, Wu W, Zhang P, Xie J. Editorial: Unveiling biomarkers and mechanisms in the tumor-immune nexus. Front Immunol 2025; 16:1581492. [PMID: 40109343 PMCID: PMC11920117 DOI: 10.3389/fimmu.2025.1581492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/22/2025] Open
Affiliation(s)
- Chenfeng Ma
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, Jiangsu, China
| | - Wantao Wu
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Pengpeng Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jiaheng Xie
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
16
|
Lu Z, Zhang C, Zhang J, Su W, Wang G, Wang Z. The Kynurenine Pathway and Indole Pathway in Tryptophan Metabolism Influence Tumor Progression. Cancer Med 2025; 14:e70703. [PMID: 40103267 PMCID: PMC11919716 DOI: 10.1002/cam4.70703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/22/2025] [Accepted: 02/04/2025] [Indexed: 03/20/2025] Open
Abstract
Tryptophan (Trp), an essential amino acid, is solely acquired through dietary intake. It is vital for protein biosynthesis and acts as a precursor for numerous key bioactive compounds. The Kynurenine Pathway and the Indole Pathway are the main metabolic routes and are extensively involved in the occurrence and progression of diseases in the digestive, nervous, and urinary systems. In the Kynurenine Pathway, enzymes crucial to tryptophan metabolism, indoleamine-2,3-dioxygenase 1 (IDO1), IDO2, and Trp-2,3-dioxygenase (TDO), trigger tumor immune resistance within the tumor microenvironment and nearby lymph nodes by depleting Trp or by activating the Aromatic Hydrocarbon Receptor (AhR) through its metabolites. Furthermore, IDO1 can influence immune responses via non-enzymatic pathways. The Kynurenine Pathway exerts its effects on tumor growth through various mechanisms, including NAD+ regulation, angiogenesis promotion, tumor metastasis enhancement, and the inhibition of tumor ferroptosis. In the Indole Pathway, indole and its related metabolites are involved in gastrointestinal homeostasis, tumor immunity, and drug resistance. The gut microbiota related to indole metabolism plays a critical role in determining the effectiveness of tumor treatment strategies and can influence the efficacy of immunochemotherapy. It is worth noting that there are conflicting effects of the Kynurenine Pathway and the Indole Pathway on the same tumor phenotype. For example, different tryptophan metabolites affect the cell cycle differently, and indole metabolism has inconsistent protective effects on tumors in different regions. These differences may hold potential for enhancing therapeutic efficacy.
Collapse
Affiliation(s)
- Zhanhui Lu
- Department of Medical Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chengcheng Zhang
- Department of Medical Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jia Zhang
- Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Wan Su
- Department of Medical Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guoying Wang
- Department of Critical Care Medicine, The Second People's Hospital of Dongying, Dongying, Shandong, China
| | - Zhongqi Wang
- Department of Medical Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
17
|
Fang P, Yang J, Zhang H, Shuai D, Li M, Chen L, Liu L. Emerging roles of intratumoral microbiota: a key to novel cancer therapies. Front Oncol 2025; 15:1506577. [PMID: 40071093 PMCID: PMC11893407 DOI: 10.3389/fonc.2025.1506577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Microorganisms, including bacteria, viruses, and fungi, have been found to play critical roles in tumor microenvironments. Due to their low biomass and other obstacles, the presence of intratumor microbes has been challenging to definitively establish. However, advances in biotechnology have enabled researchers to reveal the association between intratumor microbiota and cancer. Recent studies have shown that tumor tissues, once thought to be sterile, actually contain various microorganisms. Disrupted mucosal barriers and adjacent normal tissues are important sources of intratumor microbiota. Additionally, microbes can invade tumors by traveling through the bloodstream to the tumor site and infiltrating through damaged blood vessels. These intratumor microbiota may promote the initiation and progression of cancers by inducing genomic instability and mutations, affecting epigenetic modifications, activating oncogenic pathways, and promoting inflammatory responses. This review summarizes the latest advancements in this field, including techniques and methods for identifying and culturing intratumor microbiota, their potential sources, functions, and roles in the efficacy of immunotherapy. It explores the relationship between gut microbiota and intratumor microbiota in cancer patients, and whether altering gut microbiota might influence the characteristics of intratumor microbiota and the host immune microenvironment. Additionally, the review discusses the prospects and limitations of utilizing intratumor microbiota in antitumor immunotherapy.
Collapse
Affiliation(s)
- Pengzhong Fang
- Departments of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Jing Yang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Huiyun Zhang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Diankui Shuai
- Departments of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Min Li
- Departments of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Lin Chen
- Departments of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Liping Liu
- Departments of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
18
|
Zahedifard Z, Mahmoodi S, Ghasemian A. Genetically Engineered Bacteria as a Promising Therapeutic Strategy Against Cancer: A Comprehensive Review. Biotechnol Appl Biochem 2025. [PMID: 39985148 DOI: 10.1002/bab.2738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 02/06/2025] [Indexed: 02/24/2025]
Abstract
As a significant cause of global mortality, the cancer has also economic impacts. In the era of cancer therapy, mitigating side effects and costs and overcoming drug resistance is crucial. Microbial species can grow inside the tumor microenvironment and inhibit cancer growth through direct killing of tumor cells and immunoregulatory effects. Although microbiota or their products have demonstrated anticancer effects, the possibility of acting as pathogens and exerting side effects in certain individuals is a risk. Hence, several genetically modified/engineered bacteria (GEB) have been developed to this aim with ability of diagnosing and selective targeting and destruction of cancers. Additionally, GEB are expected to be considerably more efficient, safer, more permeable, less costly, and less invasive theranostic approaches compared to wild types. Potential GEB strains such as Escherichia coli (Nissle 1917, and MG1655), Salmonella typhimurium YB1 SL7207 (aroA gene deletion), VNP20009 (∆msbB/∆purI) and ΔppGpp (PTet and PBAD), and Listeria monocytogenes Lmat-LLO have been developed to combat cancer cells. When used in tandem with conventional treatments, GEB substantially improve the efficacy of anticancer therapy outcomes. In addition, public acceptance, optimal timing (s), duration (s), dose (s), and strains identification, interactions with other strains and the host cells, efficacy, safety and quality, and potential risks and ethical dilemmas include major challenges.
Collapse
Affiliation(s)
- Zahra Zahedifard
- Department of Medical Biotechnology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Shirin Mahmoodi
- Department of Medical Biotechnology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Abdolmajid Ghasemian
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
19
|
Wu H, Zhang W, Chang J, Wu J, Zhang X, Jia F, Li L, Liu M, Zhu J. Comprehensive analysis of mitochondrial-related gene signature for prognosis, tumor immune microenvironment evaluation, and candidate drug development in colon cancer. Sci Rep 2025; 15:6173. [PMID: 39979377 PMCID: PMC11842742 DOI: 10.1038/s41598-024-85035-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 12/30/2024] [Indexed: 02/22/2025] Open
Abstract
Colon adenocarcinoma (COAD), a common digestive system malignancy, involves crucial alterations in mitochondria-related genes influencing tumor growth, metastasis, and immune evasion. Despite limited studies on prognostic models for these genes in COAD, we established a mitochondrial-related risk prognostic model, including nine genes based on available TCGA and MitoCarta 3.0 databases, and validated its predictive power. We investigated the tumor microenvironment (TME), immune cell infiltration, complex cell communication, tumor mutation burden, and drug sensitivity of COAD patients using R language, CellChat, and additional bioinformatic tools from single-cell and bulk-tissue sequencing data. The risk model revealed significant differences in immune cell infiltration between high-risk and low-risk groups, with the strongest correlation found between tissue stem cells and macrophages in COAD. The risk score exhibited a robust correlation with TME signature genes and immune checkpoint molecules. Integrating the risk score with the immune score, microsatellite status, or TMB through TIDE analysis enhanced the accuracy of predicting immunotherapy benefits. Predicted drug efficacy offered options for both high- and low-risk group patients. Our study established a novel mitochondrial-related nine-gene prognostic signature, providing insights for prognostic assessment and clinical decision-making in COAD patients.
Collapse
Affiliation(s)
- Hao Wu
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Wentao Zhang
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Jingjia Chang
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Jin Wu
- Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Xintong Zhang
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Fengfeng Jia
- Taiyuan Technology Transfer Promotion Center, Taiyuan, 030006, China
| | - Li Li
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Ming Liu
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China.
| | - Jianjun Zhu
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
20
|
He J, Chen Y, Zhao H, Li Y. The interplay between gut bacteria and targeted therapies: implications for future cancer treatments. Mol Med 2025; 31:58. [PMID: 39948481 PMCID: PMC11827328 DOI: 10.1186/s10020-025-01108-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Targeted therapy represents a form of cancer treatment that specifically focuses on molecular markers regulating the growth, division, and dissemination of cancer cells. It serves as the cornerstone of precision medicine and is associated with fewer adverse effects compared to conventional chemotherapy, thus enhancing the quality of patient survival. These make targeted therapy as a vital component of contemporary anti-cancer strategies. Although targeted therapy has achieved excellent anti-cancer results, there are still many factors affecting its efficacy. Among the numerous factors affecting anti-cancer treatment, the role of intestinal bacteria and its metabolites are becoming increasingly prominent, particularly in immunotherapy. However, their effects on anticancer targeted therapy have not been systematically reviewed. Herein, we discuss the crosstalk between gut bacteria and anticancer targeted therapies, while also highlighting potential therapeutic strategies and future research directions.
Collapse
Affiliation(s)
- Juan He
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Yu Chen
- Department of Medical Oncology, Chongqing University Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, 400030, China
| | - Huakan Zhao
- Department of Medical Oncology, Chongqing University Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, 400030, China
| | - Yongsheng Li
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China.
- Department of Medical Oncology, Chongqing University Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, 400030, China.
| |
Collapse
|
21
|
Zhang L, Duan X, Zhao Y, Zhang D, Zhang Y. Implications of intratumoral microbiota in tumor metastasis: a special perspective of microorganisms in tumorigenesis and clinical therapeutics. Front Immunol 2025; 16:1526589. [PMID: 39995663 PMCID: PMC11847830 DOI: 10.3389/fimmu.2025.1526589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/24/2025] [Indexed: 02/26/2025] Open
Abstract
Tumor metastasis is the main cause of therapeutic failure and mortality in cancer patients. The intricate metastastic process is influenced by both the intrinsic properties of tumor cells and extrinsic factors, such as microorganisms. Notably, some microbiota have been discovered to colonize tumor tissues, collectively known as intratumoral microbiota. Intratumoral microbiota can modulate tumor progression through multiple mechanisms, including regulating immune responses, inducing genomic instability and gene mutations, altering metabolic pathways, controlling epigenetic pathways, and disrupting cancer-related signaling pathways. Furthermore, intratumoral microbiota have been shown to directly impact tumor metastasis by regulating cell adhesion, stem cell plasticity and stemness, mechanical stresses and the epithelial-mesenchymal transition. Indirectly, they may affect tumor metastasis by modulating the host immune system and the tumor microenvironment. These recent findings have reshaped our understanding of the relationship between microorganims and the metastatic process. In this review, we comprehensively summarize the existing knowledge on tumor metastasis and elaborate on the properties, origins and carcinogenic mechanisms of intratumoral microbiota. Moreover, we explore the roles of intratumoral microbiota in tumor metastasis and discuss their clinical implications. Ongoing research in this field will establish a solid foundation for novel therapeutic strategies and clinical treatments for various tumors.
Collapse
Affiliation(s)
- Lei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University,
Qingdao University, Qingdao, China
| | | | | | | | - Yuan Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University,
Qingdao University, Qingdao, China
| |
Collapse
|
22
|
Wang H, Hu J, Ma Y, Abulimiti Y, Zhou Y. Lung commensal bacteria promote lung cancer progression through NK cell-mediated immunosuppressive microenvironment. Int J Med Sci 2025; 22:1039-1051. [PMID: 40027183 PMCID: PMC11866540 DOI: 10.7150/ijms.107026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/17/2025] [Indexed: 03/05/2025] Open
Abstract
Symbiotic microbiota pervades the majority of the human body's organs and tissues, functioning as crucial regulators of both health maintenance and disease progression. Pertinently, lung adenocarcinoma has been indisputably linked to chronic inflammation. However, the precipitators that instigate such inflammation, along with the particular immune mediators involved, remain enigmatic and warrant extensive exploration. This research revealed a significant variance exists in the commensal bacteria between lung cancer tissues and their normal counterparts. This holds true for both clinical patients and mice, where both the diversity and abundance of bacteria in tumor tissues significantly surpass those in normal tissues. It has been demonstrated that disturbances in pulmonary commensal bacteria can stimulate the proliferation of tumor cells. Mechanistically, we suggest that lung bacteria may promote the expression of the NK cell immunosuppressive molecule TIGIT along with the secretion of IL-2 and IFN-γ. This consequently mediates alterations in the immunosuppressive microenvironment, thereby fostering tumor proliferation.
Collapse
Affiliation(s)
- Haiyang Wang
- Department of Laboratory Medicine, Tongji Hospital of Tongji University, School of Medicine, 389 Xincun Road, Shanghai 200065, China
| | - Jiayi Hu
- Department of Laboratory Medicine, Tongji Hospital of Tongji University, School of Medicine, 389 Xincun Road, Shanghai 200065, China
| | - Yirou Ma
- Department of Thoracic and Cardiovascular Surgery, Tongji Hospital of Tongji University, School of Medicine, 389 Xincun Road, Shanghai 200065, China
| | - Yilimunuer Abulimiti
- Department of Laboratory Medicine, Tongji Hospital of Tongji University, School of Medicine, 389 Xincun Road, Shanghai 200065, China
| | - Yongxin Zhou
- Department of Thoracic and Cardiovascular Surgery, Tongji Hospital of Tongji University, School of Medicine, 389 Xincun Road, Shanghai 200065, China
| |
Collapse
|
23
|
Jang JH, Kim DH, Chun KS. Tumor microenvironment regulation by reactive oxygen species-mediated inflammasome activation. Arch Pharm Res 2025; 48:115-131. [PMID: 39888519 DOI: 10.1007/s12272-025-01532-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025]
Abstract
Tumor microenvironment (TME) is composed of diverse cell types whose interactions, both direct and indirect, significantly influence tumorigenesis and therapeutic outcomes. Within TME, reactive oxygen species (ROS) are produced by various cells and exhibit a dual role: moderate ROS levels promote tumor initiation and progression, whereas excessive levels induce cancer cell death, influencing the efficacy of anticancer therapies. Inflammasomes, cytosolic multiprotein complexes, are pivotal in multiple stages of tumorigenesis and play a crucial role in establishing the inflammatory TME. By releasing cytokines such as IL-1β and IL-18, inflammasomes contribute to immune cell recruitment and sustain a chronic inflammatory state that supports tumor growth. ROS are critical regulators of inflammasome activation, with the impact of ROS-mediated activation differing across cell types, leading to distinct influences on tumor progression and therapeutic responses. This review explores how ROS drive inflammasome activation in various TME-associated cells and the reciprocal ROS generation induced by inflammasomes, examining their multifaceted impact on tumorigenesis and therapeutic efficacy. By elucidating the complex interplay between ROS and inflammasomes in TME, we provide insights into potential therapeutic approaches that could modulate cancer progression and enhance treatment outcomes.
Collapse
Affiliation(s)
- Jeong-Hoon Jang
- College of Pharmacy, Daegu Catholic University, Gyeongsan-si, Gyeongbuk, 38430, Republic of Korea
| | - Do-Hee Kim
- Department of Chemistry, Kyonggi University, Suwon, 16227, Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea.
| |
Collapse
|
24
|
Han S, Luo Y, Hu Z, Li X, Zhou Y, Luo F. Tumor Microenvironment Targeted by Polysaccharides in Cancer Prevention: Expanding Roles of Gut Microbiota and Metabolites. Mol Nutr Food Res 2025; 69:e202400750. [PMID: 39757562 DOI: 10.1002/mnfr.202400750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/31/2024] [Accepted: 12/02/2024] [Indexed: 01/07/2025]
Abstract
Since the development of immune checkpoint inhibitors (ICIs), immunotherapy has been widely used as a novel cancer treatment. However, the efficacy of tumor immunotherapy is largely dependent on the tumor microenvironment (TME). The high degree of heterogeneity within TME remains a major obstacle to acquire satisfactory therapeutic. Emerging studies suggest that gut microbiota is becoming an important regulator of TME. Polysaccharides as tumor immunotherapeutic agents or immune adjuvants not only exhibit antitumor activity by targeting gut microbiota, but also expand their role in the tumor immunotherapy by remodeling TME. To date, the mechanism by which polysaccharides targeting TME for tumor prevention via gut microbiota has not been deeply investigated. In this review, recent advances in the regulation of TME by polysaccharides through gut microbiota were systematically outlined, and the challenges and possible solutions in the clinical application of TME-targeted polysaccharides were discussed. Exploring the relationship between polysaccharides and TME from the perspective of gut microbiota may provide new ideas for the application of polysaccharides in tumor immunotherapy. This is a new area with major challenges that deserve further exploration.
Collapse
Affiliation(s)
- Shuai Han
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
- College of Tea and Food, Wuyi University, Wuyishan, Fujian, China
| | - Yi Luo
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zuomin Hu
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Xinhua Li
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yaping Zhou
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Feijun Luo
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
| |
Collapse
|
25
|
Yang P, Liang G, Ni Y, Chu X, Zhang X, Wang Z, Khan A, Jin F, Shen H, Li M, Xu Z. Investigating the role of intratumoral Streptococcus mitis in gastric cancer progression: insights into tumor microenvironment. J Transl Med 2025; 23:126. [PMID: 39875915 PMCID: PMC11773703 DOI: 10.1186/s12967-025-06142-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025] Open
Abstract
Growing evidence implicates that intratumoral microbiota are closely linked to cancer progression; however, research on the role of these microbiota in the development of gastric cancer remains limited. Here, using 16 S rRNA sequencing, tumor tissue proteomics and serum cytokines analysis, we identified enrichment of specific microbial communities within tumors of gastric cancer patients, possibly affecting the tumor microenvironment by immune modulation, metabolic processes, and inflammatory responses. Based on the results of in vivo experiments and intratumoral microbiota analysis, we found that Streptococcus mitis can inhibit gastric cancer progression via suppressing M2 macrophage polarization and infiltration, as well as altering the intratumoral microbial community. In summary, our findings suggest that the intratumoral microbiota, exemplified by Streptococcus mites, may be involved in regulating the progression of gastric cancer, thereby emerging as potential therapeutic targets for this disease.
Collapse
Affiliation(s)
- Ping Yang
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Physiology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, NJU Advanced Institute of Life Sciences (NAILS), Nanjing University, Nanjing, 210008, Jiangsu, P. R. China
| | - Gaoli Liang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Physiology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, NJU Advanced Institute of Life Sciences (NAILS), Nanjing University, Nanjing, 210008, Jiangsu, P. R. China
| | - Yangyue Ni
- Department of Pathogen Biology, Key Laboratory for Pathogen Infection and Control of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, Jiangsu, P.R. China
| | - Xiaojie Chu
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, 210008, China
| | - Xiaoshan Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Physiology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, NJU Advanced Institute of Life Sciences (NAILS), Nanjing University, Nanjing, 210008, Jiangsu, P. R. China
| | - Zhongyu Wang
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, 210008, China
| | - Adeel Khan
- Department of Biotechnology, University of Science and Technology Bannu, Bannu, 28100, KP, Pakistan
| | - Fangfang Jin
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China.
| | - Han Shen
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, 210008, China.
| | - Miao Li
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, 210008, China.
| | - Zhipeng Xu
- Department of Pathogen Biology, Key Laboratory for Pathogen Infection and Control of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, Jiangsu, P.R. China.
| |
Collapse
|
26
|
Li T, Zhao Z, Peng M, Zhang L, Wang C, Luo F, Zeng M, Sun K, Fang Z, Luo Y, Xie Y, Lv C, Wang J, Huang JD, Zhou H, Sun H. Multi-omics analysis reveals the interplay between intratumoral bacteria and glioma. mSystems 2025; 10:e0045724. [PMID: 39660865 PMCID: PMC11748541 DOI: 10.1128/msystems.00457-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 10/29/2024] [Indexed: 12/12/2024] Open
Abstract
Emerging evidence highlights the potential impact of intratumoral microbiota on cancer. However, the microbial composition and function in glioma remains elusive. Consequently, our study aimed to investigate the microbial community composition in glioma tissues and elucidate its role in glioma development. We parallelly performed microbial profiling, transcriptome sequencing, and metabolomics detection on tumor and adjacent normal brain tissues obtained from 50 glioma patients. We employed immunohistochemistry, multicolor immunofluorescence, and fluorescence in situ hybridization (FISH) staining to observe the presence and location of bacteria. Furthermore, an animal model was employed to validate the impact of key bacteria on glioma development. Six genera were found to be significantly enriched in glioma tissues compared to adjacent normal brain tissues, including Fusobacterium, Longibaculum, Intestinimonas, Pasteurella, Limosilactobacillus, and Arthrobacter. Both bacterial RNA and lipopolysaccharides (LPS) were observed in glioma tissues. Integrated microbiomics, transcriptomics, and metabolomics revealed that genes associated with intratumoral microbes were enriched in multiple synapse-associated pathways and that metabolites associated with intratumoral microbes were (R)-N-methylsalsolinol, N-acetylaspartylglutamic acid, and N-acetyl-l-aspartic acid. Further mediation analysis suggested that the intratumoral microbiome may affect the expression of neuron-related genes through bacteria-associated metabolites. In addition, both in vivo and in vitro models of glioma show that Fusobacterium nucleatum promotes glioma proliferation and upregulates CCL2, CXCL1, and CXCL2 levels. Our findings shed light on the intricate interplay between intratumoral bacteria and glioma. IMPORTANCE Our study adopted a multi-omics approach to unravel the impact of intratumoral microbes on neuron-related gene expression through bacteria-associated metabolites. Importantly, we found bacterial RNA and LPS signals within glioma tissues, which were traditionally considered sterile. We identified key microbiota within glioma tissues, including Fusobacterium nucleatum (Fn). Through in vivo and in vitro experiments, we identified the crucial role of Fn in promoting glioma progression, suggesting that Fn could be a potential diagnostic and therapeutic target for glioma patients. These findings offer valuable insights into the intricate interplay between intratumoral bacteria and glioma, offering novel inspiration to the realm of glioma biology.
Collapse
Affiliation(s)
- Ting Li
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhanyi Zhao
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Meichang Peng
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lu Zhang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Cheng Wang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Feiyang Luo
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Meiqin Zeng
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Kaijian Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhencheng Fang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yunhao Luo
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yugu Xie
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Cui Lv
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiaxuan Wang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian-Dong Huang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region, China
- Chinese Academy of Sciences (CAS) Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Clinical Oncology Center, Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Hongwei Zhou
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haitao Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| |
Collapse
|
27
|
Liu G, Liu K, Ji L, Li Y. Intratumoral microbiota, fatty acid metabolism, and tumor microenvironment constitute an unresolved trinity in colon adenocarcinoma. Sci Rep 2025; 15:2568. [PMID: 39833403 PMCID: PMC11747563 DOI: 10.1038/s41598-025-87194-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025] Open
Abstract
The intratumoral microbiota, fatty acid metabolism (FAM), and tumor microenvironment (TME) all provide insights into the management of colon adenocarcinoma (COAD). But the biological link among the three remains unclear. Here, we analyzed intratumoral microbiome samples and matched host transcriptome samples from 420 patients with COAD in The Cancer Genome Atlas (TCGA). All patients were divided into two subtypes (FAM_high and FAM_low) based on the Gene set variation analysis (GSVA) score of FAM pathway. Furthermore, we found significant difference in the intratumoral microbiota signatures between the two subtypes. In-depth analysis suggested that specific microbes in tumors may indirectly modify the TME, particularly stromal cell populations, by modulating the FAM process. More importantly, the crosstalk between the three can have a significant impact on prognosis, response to immunotherapy, and drug sensitivity of patients. Pathological image profiling showed that changes in the TME originating from intratumoral microbiota disturbance could be reflected in pathological image features. In summary, our study provides novel insights into the biological links among the intratumoral microbiota, FAM, and the TME in COAD, and offer guidance for the therapeutic opportunities that target intratumoral microbes.
Collapse
Affiliation(s)
- Guangyi Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kun Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lei Ji
- Geneis Beijing Co., Ltd., Beijing, 100102, China
| | - Yang Li
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
28
|
Shu F, Yu J, Liu Y, Wang F, Gou G, Wen M, Luo C, Lu X, Hu Y, Du Q, Xu J, Xie R. Mast cells: key players in digestive system tumors and their interactions with immune cells. Cell Death Discov 2025; 11:8. [PMID: 39814702 PMCID: PMC11735678 DOI: 10.1038/s41420-024-02258-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/22/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025] Open
Abstract
Mast cells (MCs) are critical components of both innate and adaptive immune processes. They play a significant role in protecting human health and in the pathophysiology of various illnesses, including allergies, cardiovascular diseases and autoimmune diseases. Recent studies in tumor-related research have demonstrated that mast cells exert a substantial influence on tumor cell behavior and the tumor microenvironment, exhibiting both pro- and anti-tumor effects. Specifically, mast cells not only secrete mediators related to pro-tumor function such as trypsin-like enzymes, chymotrypsin, vascular endothelial cell growth factor and histamine, but also mediators related to anti-tumor progression such as cystatin C and IL-17F. This dual role of mast cells renders them an under-recognized but very promising target for tumor immunotherapy. Digestive system tumors, characterized by high morbidity and associated mortality rates globally, are increasingly recognized as a significant healthcare burden. This paper examines the influence of mast cell-derived mediators on the development of tumors in the digestive system. It also explores the prognostic significance of mast cells in patients with various gastrointestinal cancers at different stages of the disease. Additionally, the article investigates the interactions between mast cells and immune cells, as well as the potential relationships among intratumoral bacteria, immune cells, and mast cell within digestive system microenvironment. The aim is to propose new strategies for the immunotherapy of digestive system tumors by targeting mast cells.
Collapse
Affiliation(s)
- Feihong Shu
- Department of Endoscopy and Digestive System, Guizhou Provincial People's Hospital, Guiyang, China
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Jie Yu
- Department of Endoscopy and Digestive System, Guizhou Provincial People's Hospital, Guiyang, China
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Youjia Liu
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Fang Wang
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Guoyou Gou
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Min Wen
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Chen Luo
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Xianmin Lu
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Yanxia Hu
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Qian Du
- Department of Endoscopy and Digestive System, Guizhou Provincial People's Hospital, Guiyang, China
| | - Jingyu Xu
- Guizhou Medical University, Guiyang, Guizhou, China
| | - Rui Xie
- Department of Endoscopy and Digestive System, Guizhou Provincial People's Hospital, Guiyang, China.
| |
Collapse
|
29
|
Qin G, Shao X, Liu X, Xu J, Wang X, Wang W, Gao L, Liang Y, Xie L, Su D, Yang H, Zhou W, Fang X. A signaling molecule from intratumor bacteria promotes trastuzumab resistance in breast cancer cells. Proc Natl Acad Sci U S A 2025; 122:e2421710122. [PMID: 39786928 PMCID: PMC11745319 DOI: 10.1073/pnas.2421710122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/02/2024] [Indexed: 01/12/2025] Open
Abstract
Emerging evidence indicates that intratumor bacteria exist as an active and specific tumor component in many tumor types beyond digestive and respiratory tumors. However, the biological impact and responsible molecules of such local bacteria-tumor direct interaction on cancer therapeutic response remain poorly understood. Trastuzumab is among the most commonly used drugs targeting the receptor tyrosine-protein kinase erbB-2 (ErbB2) in breast cancer, but its resistance is inevitable, severely limiting its clinical effectiveness. Here, we demonstrate that the quorum-sensing signaling molecule N-(3-oxo-dodecanoyl) homoserine lactone (3oc), a chemical compound released by Pseudomonas aeruginosa (P. aeruginosa), one tumor-resident bacteria with a relative high abundance in breast cancer, promotes breast cancer cell resistance to trastuzumab. Mechanically, 3oc directly leads to spontaneous dimerization of the transforming growth factor β (TGF-β) type II serine/threonine kinase receptor on the cell membrane in a ligand-independent manner. The 3oc-induced TGF-β signaling subsequently triggers ErbB2 phosphorylation and its downstream target activation, overcoming the inhibition effect of trastuzumab on ErbB2. With specific real-time qPCR, fluorescence in situ hybridization imaging, and liquid chromatography ionization tandem mass spectrometry analyses of clinical samples, we confirmed that P. aeruginosa and its signaling molecule 3oc exist in breast cancer tissues and there is a clinical correlation between P. aeruginosa colonization and trastuzumab resistance. This work expands the biological functions of intratumor bacteria in cancer treatment responsiveness and provides a unique perspective for overcoming trastuzumab resistance.
Collapse
Affiliation(s)
- Gege Qin
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing100190, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, Zhejiang, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua-Peking Center for Life Sciences, Institute of Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing100084, China
| | - Xiying Shao
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou310022, Zhejiang, China
| | - Xiaolong Liu
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing100190, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Jiachao Xu
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing100101, China
| | - Xiaojia Wang
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou310022, Zhejiang, China
| | - Wenxi Wang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, Zhejiang, China
| | - Lu Gao
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, Zhejiang, China
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou310022, Zhejiang, China
| | - Yuxin Liang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing100190, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Lina Xie
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou310022, Zhejiang, China
| | - Dan Su
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou310022, Zhejiang, China
| | - Hongwei Yang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing100190, China
| | - Wei Zhou
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, Zhejiang, China
| | - Xiaohong Fang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing100190, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, Zhejiang, China
- University of Chinese Academy of Sciences, Beijing100049, China
| |
Collapse
|
30
|
Wawrety W, Kedziora ,A. Role of bacteria in cancers and their therapeutic potential: Review of current knowledge. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2025; 28:273-282. [PMID: 39906620 PMCID: PMC11790194 DOI: 10.22038/ijbms.2024.77667.16798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 05/14/2024] [Indexed: 02/06/2025]
Abstract
Cancers are extremely dynamic diseases that can actively cause refractorines to be gained from applied therapies, which is why they are at the forefront of deaths worldwide. In this literature review, we covered the most recent and important discoveries regarding the influence of human microbiota, including tumor bacteriome, on the development and treatment of cancer. Advances in research on microbial communities have enabled us to discover the role of the human microbiome in the development and course of this disease, helping us understand neoplasms better and design new potential therapies. As we show through our findings, by immunomodulation and the secretion of certain chemical substances, the correct bacteriome of the intestinal tract, respiratory system, or skin can protect humans against cancer development and help during the treatment process. Bacteria also reside inside tumors, forming part of the tumor microenvironment (TME), where they interact with immunological and cancer cells in many complex ways. Some bacteria, such as Pseudomonas aeruginosa or Akkermansia muciniphila, can stimulate anticancer cell-mediated immune responses or even directly lead to cancer cell death. We also present the clinical possibilities of using some live, usually modified bacteria to develop bacteriotherapies. Modifying the gut microbiome to stimulate standard treatment is also important. Research on the microbiome and cancer remains a challenging topic in microbiology, having a great potential for advancements in cancer therapy in the future, and is continuously becoming a more and more popular field of research, as shown by our statistical analysis of PubMed data.
Collapse
Affiliation(s)
- Wojciech Wawrety
- Department of Microbiology Faculty of Biological Sciences University of Wroclaw Przybyszewskiego 63, 51-148 Wroclaw, Poland
| | - , Anna Kedziora
- Department of Microbiology Faculty of Biological Sciences University of Wroclaw Przybyszewskiego 63, 51-148 Wroclaw, Poland
| |
Collapse
|
31
|
D’Antonio DL, Zenoniani A, Umme S, Piattelli A, Curia MC. Intratumoral Fusobacterium nucleatum in Pancreatic Cancer: Current and Future Perspectives. Pathogens 2024; 14:2. [PMID: 39860963 PMCID: PMC11768203 DOI: 10.3390/pathogens14010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
The intratumoral microbiome plays a significant role in many cancers, such as lung, pancreatic, and colorectal cancer. Pancreatic cancer (PC) is one of the most lethal malignancies and is often diagnosed at advanced stages. Fusobacterium nucleatum (Fn), an anaerobic Gram-negative bacterium primarily residing in the oral cavity, has garnered significant attention for its emerging role in several extra-oral human diseases and, lately, in pancreatic cancer progression and prognosis. It is now recognized as oncobacterium. Fn engages in pancreatic tumorigenesis and metastasis through multifaceted mechanisms, including immune response modulation, virulence factors, control of cell proliferation, intestinal metabolite interactions, DNA damage, and epithelial-mesenchymal transition. Additionally, compelling research suggests that Fn may exert detrimental effects on cancer treatment outcomes. This paper extends the perspective to pancreatic cancer associated with Fn. The central focus is to unravel the oncogenomic changes driven by Fn in colonization, initiation, and promotion of pancreatic cancer development. The presence of Fusobacterium species can be considered a prognostic marker of PC, and it is also correlated to chemoresistance. Furthermore, this review underscores the clinical research significance of Fn as a potential tumor biomarker and therapeutic target, offering a novel outlook on its applicability in cancer detection and prognostic assessment. It is thought that given the role of Fn in tumor formation and metastasis processes via its FadA, FapA, Fap2, and RadD, new therapies for tumor treatment targeting Fn will be developed.
Collapse
Affiliation(s)
- Domenica Lucia D’Antonio
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (D.L.D.); (A.Z.); (S.U.)
| | - Anna Zenoniani
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (D.L.D.); (A.Z.); (S.U.)
| | - Samia Umme
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (D.L.D.); (A.Z.); (S.U.)
- Department of Neuroscience, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy
| | - Adriano Piattelli
- School of Dentistry, Saint Camillus International University of Health and Medical Sciences (UniCamillus), 00131 Rome, Italy;
- Facultad de Medicina, UCAM Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain
| | - Maria Cristina Curia
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (D.L.D.); (A.Z.); (S.U.)
| |
Collapse
|
32
|
Jin L, Lin Z, Jin A. Sterol regulatory element binding transcription factor 1 is an important prognostic factor for colon adenocarcinoma and closely related to immune infiltration. Cytojournal 2024; 21:67. [PMID: 39917010 PMCID: PMC11801665 DOI: 10.25259/cytojournal_43_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 11/18/2024] [Indexed: 02/09/2025] Open
Abstract
Objective Sterol Regulatory Element Binding Transcription Factor 1 (SREBF1) encodes a core protein that has a crucial function in the metabolism of cholesterol and lipids. This transcription factor is a member of the family of transcription factors and highly expressed in a variety of cancer types. As of now, there are few reports on the relationship between the expression of SREBF1 and colon adenocarcinoma (COAD). Hence, this study utilizes databases and a range of experiments to explore the relationship between the expression of SREBF1 and tumor immune infiltration, as well as the occurrence and development of tumors. Material and Methods The expression of SREBF1 in pan-cancers was retrieved through databases such as TIMER, Gene Expression Profiling Interactive Analysis (GEPIA), and UALCAN. The expression of SREBF1 in HCT-116 and SW480 cells was detected using western blot. Furthermore, we also found that knockdown SREBF1 can inhibit the proliferation and migration of COAD cells. The correlation between SREBF1 and autophagy in COAD cells was detected using acridine orange (AO) staining, western blot, and immunofluorescence (IF). Results The databases of TIMER, GEPIA and UALCAN revealed that SREBF1 is overexpressed in pan-cancer tissues, and closely associated with the prognosis of the patients with cancer. Further immunohistochemical staining showed that SREBF1 was overexpressed in COAD, and closely related to the clinical stage and lymph node metastasis. Western blot revealed that SREBF1 was significantly expressed in both HCT-116 and SW480 COAD cells; knockdown of SREBF1 could inhibit the proliferation, DNA replication, and migration of COAD cells. The AO staining, western blot, and IF experiments also showed that silencing SREBF1 could promote the autophagy of COAD cell. Meanwhile, the TIMER database indicates a significant positive correlation between the presence of immune cells in COAD and variations in copy number alteration of SREBF1. Conclusion SREBF1 might serve as a potential prognostic marker for COAD and be associated with immune cell infiltration.
Collapse
Affiliation(s)
- Liying Jin
- Central Laboratory, The Affiliated Hospital of Yanbian University, Yanji, Jilin, China
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, Jilin, China
| | - Zhenhua Lin
- Central Laboratory, The Affiliated Hospital of Yanbian University, Yanji, Jilin, China
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, Jilin, China
| | - Aihua Jin
- Central Laboratory, The Affiliated Hospital of Yanbian University, Yanji, Jilin, China
| |
Collapse
|
33
|
Huang Y, Zhang L, Zhang W, Lv N, Wang T. Diagnostic and prognostic values of NSCLC patients with or without obstructive pneumonia after sleeve lobectomy. Front Cell Infect Microbiol 2024; 14:1474998. [PMID: 39735257 PMCID: PMC11682716 DOI: 10.3389/fcimb.2024.1474998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
Objective We aimed to identify the diagnostic value of next-generation sequencing (NGS) of bronchoalveolar lavage fluid (BALF) from patients with non-small-cell lung cancer (NSCLC). Methods Forty patients who were initially diagnosed with pulmonary nodules were enrolled. Frozen section histology was used to identify the NSCLC cell types. NGS of collected BALF samples was used for microbial identification. We compared the bacterial and viral distributions in BALF samples from patients with NSCLC with and without obstructive pneumonia as well as their NSCLC drainage times following surgery. Results Of the 29 patients with NSCLC, eight had obstructive pneumonia. Streptococcus pneumoniae, Streptococcus pseudopneumoniae, and Haemophilus parainfluenzae were the top three bacteria present in almost 50% of patients, both with and without obstructive pneumonia. The viral detection rate was higher in the BALF of patients with NSCLC who did not have obstructive pneumonia. However, in patients with NSCLC and drain times of >5 days, the human herpes virus type 7 detection rate was higher following surgery than it was in patients with NSCLC who had drain times of ≤5 days. Conclusion Viral imbalance in NSCLC is closely related to the occurrence of obstructive pneumonia and postoperative drainage time.
Collapse
Affiliation(s)
- Yuxia Huang
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lan Zhang
- Department of Operating Room, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wentian Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Na Lv
- Department of Operating Room, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tao Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
34
|
Wang J, Li H, Wang Z, Ruan S. Luteolin: A Comprehensive and Visualized Analysis of Research Hotspots and Its Antitumor Mechanisms. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:2377-2401. [PMID: 39686791 DOI: 10.1142/s0192415x24500903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
The aim of this study was to analyze the research hotspots and mechanisms of luteolin in tumor-related fields using bibliometric and bioinformatic approaches to guide future research. We conducted a comprehensive screening of all articles on luteolin and tumors in Web of Science from 2008 to 2023. The extracted words from these publications were visualized using VOSviewer, Scimago Graphica, and CiteSpace. Public databases were used to collect luteolin and tumor-related targets. GO and KEGG analyses of luteolin antitumor-related genes were performed using Metascape. Protein interaction networks were built with Cytoscape and STRING, identifying hub targets of luteolin in antitumor activity. Subsequently, the binding capacity of luteolin to these hub targets was assessed using molecular docking technology. We found that China dominated this field, the Egyptian Knowledge Bank published the most articles, and Molecules had the highest number of related publications. Recently, network pharmacology, target, traditional Chinese medicine, and nanoparticles have become research hotspots in luteolin's antitumor research. A total of 483 overlapping genes between luteolin and tumors were identified, and they were closely associated with the cancer-associated pathways, PI3K-Akt, and MAPK signaling pathways. Luteolin forms a complex network of antitumor-related genes, with TP53, TNF, STAT3, AKT1, JUN, IL6, and SRC playing key roles and showing strong binding affinity to luteolin. Computer technology is becoming increasingly integral to the discipline, and future research will focus on more precise antitumor mechanisms and effective clinical applications.
Collapse
Affiliation(s)
- Jiaxuan Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, P. R. China
| | - Hao Li
- The First Affiliated Hospital of Zhejiang Chinese Medical University, (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, P. R. China
| | - Zhenru Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, P. R. China
| | - Shanming Ruan
- The First Affiliated Hospital of Zhejiang Chinese Medical University, (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, P. R. China
| |
Collapse
|
35
|
Zhang Z, Wu W, Lin J, Li H. Unveiling the hidden causal links: skin flora and cutaneous melanoma. Front Oncol 2024; 14:1451175. [PMID: 39723372 PMCID: PMC11668787 DOI: 10.3389/fonc.2024.1451175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Objective The presence of skin flora (SF) has been identified as a significant factor in the onset and progression of cutaneous melanoma (CM). However, the vast diversity and abundance of SF present challenges to fully understanding the causal relationship between SF and CM. Methods A Two Sample Mendelian Randomization (TSMR) analysis was conducted to investigating the causal relationship between SF and CM. The Inverse-Variance Weighted (IVW) method was utilized as the primary approach to assess the causal relationship under investigation. Furthermore, an independent external cohort was employed to validate the initial findings, followed by a meta-analysis of the consolidated results. To address potential confounding factors related to the influence of SF on CM, a Multivariate Mendelian Randomization (MVMR) analysis was also conducted. Finally, a Reverse Mendelian Randomization (RMR) was conducted to further validate the causal association. Results TSMR results showed that 9 SF have a causal relationship with CM in the training cohort. Although these 9 SF weren't confirmed in the testing cohort, 4 SF remained significant in the meta-analysis after integrating results from both cohorts. MVMR analysis indicated that 3 SF were still significantly associated with CM after accounting for the interactions between different SF in the training cohort. No reverse causal relationship was identified in RMR analysis. Conclusion A total of 9 SF were identified as having a potential causal relationship with CM; however, a large randomized controlled trial is needed to verify these results.
Collapse
Affiliation(s)
- Zexin Zhang
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenfeng Wu
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiajia Lin
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongyi Li
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
36
|
Zou J, Xu B, Gao H, Luo P, Chen T, Duan H. Microbiome in urologic neoplasms: focusing on tumor immunity. Front Immunol 2024; 15:1507355. [PMID: 39703512 PMCID: PMC11655508 DOI: 10.3389/fimmu.2024.1507355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/18/2024] [Indexed: 12/21/2024] Open
Abstract
Urological tumors are an important disease affecting global human health, and their pathogenesis and treatment have been the focus of medical research. With the in - depth study of microbiomics, the role of the microbiome in urological tumors has gradually attracted attention. However, the current research on tumor - associated microorganisms mostly focuses on one type or one site, and currently, there is a lack of attention to the microbiome in the immunity and immunotherapy of urological tumors. Therefore, in this paper, we systematically review the distribution characteristics of the microbiome (including microorganisms in the gut, urine, and tumor tissues) in urologic tumors, the relationship with disease prognosis, and the potential mechanisms of microbial roles in immunotherapy. In particular, we focus on the molecular mechanisms by which the microbiome at different sites influences tumor immunity through multiple "messengers" and pathways. We aim to further deepen the understanding of microbiome mechanisms in urologic tumors, and also point out the direction for the future development of immunotherapy for urologic tumors.
Collapse
Affiliation(s)
- Jun Zou
- Department of Otorhinolaryngology, The Affiliated Fengcheng Hospital of Yichun University, Fengcheng, Jiangxi, China
| | - Baisheng Xu
- Department of Urology, The First People's Hospital of Xiushui, Jiujiang, Jiangxi, China
| | - Hongbing Gao
- Department of Urology, The First People's Hospital of Xiushui, Jiujiang, Jiangxi, China
| | - Peiyue Luo
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Tao Chen
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Huanglin Duan
- Department of Urology, The First People's Hospital of Xiushui, Jiujiang, Jiangxi, China
| |
Collapse
|
37
|
Ma Y, Chen T, Sun T, Dilimulati D, Xiao Y. The oncomicrobiome: New insights into microorganisms in cancer. Microb Pathog 2024; 197:107091. [PMID: 39481695 DOI: 10.1016/j.micpath.2024.107091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/15/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
The discoveries of the oncomicrobiome (intratumoral microbiome) and oncomicrobiota (intratumoral microbiota) represent significant advances in tumor research and have rapidly become of key interest to the field. Within tumors, microorganisms such as bacteria, fungi, viruses, and archaea form the oncomicrobiota and are primarily found within tumor cells, immunocytes, and the intercellular matrix. The oncomicrobiome exhibits marked heterogeneity and is associated with tumor initiation, progression, metastasis, and treatment response. Interactions between the oncomicrobiome and the immune system can modulate host antitumor immunity, influencing the efficacy of immunotherapies. Oncomicrobiome research also faces numerous challenges, including overcoming methodological issues such as low target abundance, susceptibility to contamination, and biases in sample handling and analysis methods across different studies. Furthermore, studies of the oncomicrobiome may be confounded by baseline differences in microbiomes among populations driven by both environmental and genetic factors. Most studies to date have revealed associations between the oncomicrobiome and tumors, but very few have established mechanistic links between the two. This review introduces the relevant concepts, detection methods, sources, and characteristics of the oncomicrobiome. We then describe the composition of the oncomicrobiome in common tumors and its role in shaping the tumor microenvironment. We also discuss the current problems and challenges to be overcome in this rapidly progressing field.
Collapse
Affiliation(s)
- Yingying Ma
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tingting Sun
- Department of Structure and Morphology, Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China; Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, China
| | - Dilinuer Dilimulati
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yonghong Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Structure and Morphology, Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China; Peking Union Medical College & Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
38
|
Li J, Li Y, Zhou L, Li H, Wan T, Tang J, Zhou L, Xie H, Wang L. Microbiome analysis reveals the inducing effect of Pseudomonas on prostatic hyperplasia via activating NF-κB signalling. Virulence 2024; 15:2313410. [PMID: 38378443 PMCID: PMC10880505 DOI: 10.1080/21505594.2024.2313410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/25/2024] [Indexed: 02/22/2024] Open
Abstract
Benign prostatic hyperplasia (BPH) is a prevalent disease among middle-aged and elderly males, but its pathogenesis remains unclear. Dysbiosis of the microbiome is increasingly recognized as a significant factor in various human diseases. Prostate tissue also contains a unique microbiome, and its dysbiosis has been proposed to contribute to prostate diseases. Here, we obtained prostate tissues and preoperative catheterized urine from 24 BPH individuals, and 8 normal prostate samples as controls, which followed strict aseptic measures. Using metagenomic next-generation sequencing (mNGS), we found the disparities in the microbiome composition between normal and BPH tissues, with Pseudomonas significantly enriched in BPH tissues, as confirmed by fluorescence in situ hybridization (FISH). Additionally, we showed that the prostate microbiome differed from the urine microbiome. In vitro experiments revealed that lipopolysaccharide (LPS) of Pseudomonas activated NF-κB signalling, leading to inflammation, proliferation, and EMT processes, while inhibiting apoptosis in prostatic cells. Overall, our research determines the presence of microbiome dysbiosis in BPH, and suggests that Pseudomonas, as the dominant microflora, may promote the progression of BPH through LPS activation of NF-κB signalling.
Collapse
Affiliation(s)
- Jiaren Li
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Youyou Li
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liang Zhou
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongming Li
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tengfei Wan
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jin Tang
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Zhou
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hui Xie
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Long Wang
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
39
|
Harmak Z, Kone AS, Ghouzlani A, Ghazi B, Badou A. Beyond Tumor Borders: Intratumoral Microbiome Effects on Tumor Behavior and Therapeutic Responses. Immune Netw 2024; 24:e40. [PMID: 39801738 PMCID: PMC11711125 DOI: 10.4110/in.2024.24.e40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 01/16/2025] Open
Abstract
The human body contains a diverse array of microorganisms, which exert a significant impact on various physiological processes, including immunity, and can significantly influence susceptibility to various diseases such as cancer. Recent advancements in metagenomic sequencing have uncovered the role of intratumoral microbiome, which covertly altered the development of cancer, the growth of tumors, and the response to existing treatments through multiple mechanisms. These mechanisms involve mainly DNA damage induction, oncogenic signaling pathway activation, and the host's immune response modulation. To explore novel therapeutic options and effectively target and regulate the intratumoral microbiome, a comprehensive understanding of these processes is indispensable. Here, we will explore various potential actions of the intratumoral microbiome concerning the initiation and progression of tumors. We will examine its impact on responses to chemotherapy, radiotherapy, and immunotherapy. Additionally, we will discuss the current state of knowledge regarding the use of genetically modified bacteria as a promising treatment option for cancer.
Collapse
Affiliation(s)
- Zakia Harmak
- Immuno-Genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, University Hassan II, Casablanca 20000, Morocco
| | - Abdou-Samad Kone
- Immuno-Genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, University Hassan II, Casablanca 20000, Morocco
| | - Amina Ghouzlani
- Immuno-Genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, University Hassan II, Casablanca 20000, Morocco
| | - Bouchra Ghazi
- Immunopathology-Immunomonitoring-Immunotherapy Laboratory, Faculty of Medicine, Mohammed IV University of Sciences and Health, Casablanca 82403, Morocco
- IVF Laboratory, Department of Reproductive Medicine, Mohammed VI International University Hospital, Bouskoura 27182, Morocco
| | - Abdallah Badou
- Immuno-Genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, University Hassan II, Casablanca 20000, Morocco
| |
Collapse
|
40
|
Thapa R, Magar AT, Shrestha J, Panth N, Idrees S, Sadaf T, Bashyal S, Elwakil BH, Sugandhi VV, Rojekar S, Nikhate R, Gupta G, Singh SK, Dua K, Hansbro PM, Paudel KR. Influence of gut and lung dysbiosis on lung cancer progression and their modulation as promising therapeutic targets: a comprehensive review. MedComm (Beijing) 2024; 5:e70018. [PMID: 39584048 PMCID: PMC11586092 DOI: 10.1002/mco2.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/26/2024] Open
Abstract
Lung cancer (LC) continues to pose the highest mortality and exhibits a common prevalence among all types of cancer. The genetic interaction between human eukaryotes and microbial cells plays a vital role in orchestrating every physiological activity of the host. The dynamic crosstalk between gut and lung microbiomes and the gut-lung axis communication network has been widely accepted as promising factors influencing LC progression. The advent of the 16s rDNA sequencing technique has opened new horizons for elucidating the lung microbiome and its potential pathophysiological role in LC and other infectious lung diseases using a molecular approach. Numerous studies have reported the direct involvement of the host microbiome in lung tumorigenesis processes and their impact on current treatment strategies such as radiotherapy, chemotherapy, or immunotherapy. The genetic and metabolomic cross-interaction, microbiome-dependent host immune modulation, and the close association between microbiota composition and treatment outcomes strongly suggest that designing microbiome-based treatment strategies and investigating new molecules targeting the common holobiome could offer potential alternatives to develop effective therapeutic principles for LC treatment. This review aims to highlight the interaction between the host and microbiome in LC progression and the possibility of manipulating altered microbiome ecology as therapeutic targets.
Collapse
Affiliation(s)
- Rajan Thapa
- Department of Pharmacy, Universal college of medical sciencesTribhuvan UniversityBhairahawaRupendehiNepal
| | - Anjana Thapa Magar
- Department of MedicineKathmandu Medical College Teaching Hospital, SinamangalKathmanduNepal
| | - Jesus Shrestha
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNew South WalesAustralia
| | - Nisha Panth
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Sobia Idrees
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Tayyaba Sadaf
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Saroj Bashyal
- Department of Pharmacy, Manmohan Memorial Institute of Health SciencesTribhuvan University, SoalteemodeKathmanduNepal
| | - Bassma H. Elwakil
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences TechnologyPharos University in AlexandriaAlexandriaEgypt
| | - Vrashabh V. Sugandhi
- Department of pharmaceutical sciences, College of Pharmacy & Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - Satish Rojekar
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ram Nikhate
- Department of PharmaceuticsDattakala Shikshan Sanstha, Dattakala college of pharmacy (Affiliated to Savitribai Phule Pune universityPuneMaharashtraIndia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical SciencesSaveetha UniversityChennaiIndia
- Centre of Medical and Bio‐allied Health Sciences ResearchAjman UniversityAjmanUAE
| | - Sachin Kumar Singh
- School of Pharmaceutical SciencesLovely Professional UniversityPhagwaraIndia
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
- Discipline of Pharmacy, Graduate School of HealthUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Philip M Hansbro
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| |
Collapse
|
41
|
Yang M, Lin W, Huang J, Mannucci A, Luo H. Novel immunotherapeutic approaches in gastric cancer. PRECISION CLINICAL MEDICINE 2024; 7:pbae020. [PMID: 39397869 PMCID: PMC11467695 DOI: 10.1093/pcmedi/pbae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/08/2024] [Accepted: 09/08/2024] [Indexed: 10/15/2024] Open
Abstract
Gastric cancer is a malignant tumor that ranks third in cancer-related deaths worldwide. Early-stage gastric cancer can often be effectively managed through surgical resection. However, the majority of cases are diagnosed in advanced stages, where outcomes with conventional radiotherapy and chemotherapy remain unsatisfactory. Immunotherapy offers a novel approach to treating molecularly heterogeneous gastric cancer by modifying the immunosuppressive tumor microenvironment. Immune checkpoint inhibitors and adoptive cell therapy are regarded as promising modalities in cancer immunotherapy. Food and Drug Administration-approved programmed death-receptor inhibitors, such as pembrolizumab, in combination with chemotherapy, have significantly extended overall survival in gastric cancer patients and is recommended as a first-line treatment. Despite challenges in solid tumor applications, adoptive cell therapy has demonstrated efficacy against various targets in gastric cancer treatment. Among these approaches, chimeric antigen receptor-T cell therapy research is the most widely explored and chimeric antigen receptor-T cell therapy targeting claudin18.2 has shown acceptable safety and robust anti-tumor capabilities. However, these advancements primarily remain in preclinical stages and further investigation should be made to promote their clinical application. This review summarizes the latest research on immune checkpoint inhibitors and adoptive cell therapy and their limitations, as well as the role of nanoparticles in enhancing immunotherapy.
Collapse
Affiliation(s)
- Meng Yang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Wuhao Lin
- Department of Molecular Diagnostics, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jiaqian Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Alessandro Mannucci
- Gastroenterology and Gastrointestinal Emndoscopy Unit, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan 20132, Italy
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope; Monrovia, CA 91016, USA
| | - Huiyan Luo
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| |
Collapse
|
42
|
Ciernikova S, Sevcikova A, Mego M. Targeting the gut and tumor microbiome in cancer treatment resistance. Am J Physiol Cell Physiol 2024; 327:C1433-C1450. [PMID: 39437444 DOI: 10.1152/ajpcell.00201.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Therapy resistance represents a significant challenge in oncology, occurring in various therapeutic approaches. Recently, animal models and an increasing set of clinical trials highlight the crucial impact of the gut and tumor microbiome on treatment response. The intestinal microbiome contributes to cancer initiation, progression, and formation of distant metastasis. In addition, tumor-associated microbiota is considered a critical player in influencing tumor microenvironments and regulating local immune processes. Intriguingly, numerous studies have successfully identified pathogens within the gut and tumor microbiome that might be linked to a poor response to different therapeutic modalities. The unfavorable microbial composition with the presence of specific microbes participates in cancer resistance and progression via several mechanisms, including upregulation of oncogenic pathways, macrophage polarization reprogramming, metabolism of chemotherapeutic compounds, autophagy pathway modulation, enhanced DNA damage repair, inactivation of a proapoptotic cascade, and bacterial secretion of extracellular vesicles, promoting the processes in the metastatic cascade. Targeted elimination of specific intratumoral bacteria appears to enhance treatment response. However, broad-spectrum antibiotic pretreatment is mostly connected to reduced efficacy due to gut dysbiosis and lower diversity. Mounting evidence supports the potential of microbiota modulation by probiotics and fecal microbiota transplantation to improve intestinal dysbiosis and increase microbial diversity, leading to enhanced treatment efficacy while mitigating adverse effects. In this context, further research concerning the identification of clinically relevant microbiome signatures followed by microbiota-targeted strategies presents a promising approach to overcoming immunotherapy and chemotherapy resistance in refractory patients, improving their outcomes.
Collapse
Affiliation(s)
- Sona Ciernikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Aneta Sevcikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Michal Mego
- 2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| |
Collapse
|
43
|
Hong BY, Chhaya A, Robles A, Cervantes J, Tiwari S. The role of Fusobacterium nucleatum in the pathogenesis of colon cancer. J Investig Med 2024; 72:819-827. [PMID: 39175147 DOI: 10.1177/10815589241277829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Previously, many studies have reported changes in the gut microbiota of patients with colorectal cancer (CRC). While CRC is a well-described disease, the relationship between its development and features of the intestinal microbiome is still being understood. Evidence linking Fusobacterium nucleatum enrichment in colorectal tumor tissue has prompted the elucidation of various molecular mechanisms and tumor-promoting attributes. In this review we highlight various aspects of our understanding of the relationship between the development of CRC and the alteration of intestinal microbiome, focusing specifically on the role of F. nucleatum. As the amount of F. nucleatum DNA in CRC tissue is associated with shorter survival, it may potentially serve as a prognostic biomarker, and most importantly may open the door for a role in CRC treatment.
Collapse
Affiliation(s)
- Bo-Young Hong
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Ajay Chhaya
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA
| | - Alejandro Robles
- Department of Internal Medicine, Division of Gastroenterology, Texas Tech University Health Sciences Center El Paso, El Paso, TX, USA
| | - Jorge Cervantes
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Sangeeta Tiwari
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA
- Biomedical Research Center, University of Texas at El Paso, El Paso, TX, USA
| |
Collapse
|
44
|
Saadh MJ, Ahmed HH, Al-Hussainy AF, Kaur I, Kumar A, Chahar M, Saini S, Taher WM, Alwan M, Jawad MJ, Darvishi M, Alsaikhan F. Bile's Hidden Weapon: Modulating the Microbiome and Tumor Microenvironment. Curr Microbiol 2024; 82:25. [PMID: 39614901 DOI: 10.1007/s00284-024-04004-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024]
Abstract
The human gut microbiome is a dynamic and intricate ecosystem, composed of trillions of microorganisms that play a pivotal role in maintaining overall health and well-being. However, the gut microbiome is constantly exposed to various environmental factors, including the bile produced by the liver, which can significantly impact its composition and function. Bile acids, secreted by the liver and stored in the gallbladder, modulate the gut microbiome, influencing its composition and function. This altered microbiome profile can, in turn, impact the tumor microenvironment (TME), promoting an immunosuppressive environment that favors tumor growth and metastasis. Furthermore, changes in the gut microbiome can also influence the production of bile acids and other metabolites that directly affect cancer cells and their behavior. Moreover, bile acids have been shown to shape the microbiome and increase antibiotic resistance, underscoring the need for targeted interventions. This review provides a comprehensive overview of the intricate relationships between bile, the gut microbiome, and the TME, highlighting the mechanisms by which this interplay drives cancer progression and resistance to therapy. Understanding these complex interactions is crucial for developing novel therapeutic strategies that target the gut-bile-TME axis and improve patient outcomes.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | | | - Irwanjot Kaur
- Department of Biotechnology and Genetics, Jain (Deemed-to-Be) University, Bengaluru, Karnataka, 560069, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Abhishek Kumar
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh, 247341, India
- Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand, 831001, India
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering & Technology, NIMS University, Rajasthan, Jaipur, India
| | - Suman Saini
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | - Mohammad Darvishi
- Infectious Diseases and Tropical Medicine Research Center (IDTMRC), Department of Aerospace and Subaquatic Medicine, AJA University of Medical Sciences, Tehran, Iran.
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
- School of Pharmacy, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia
| |
Collapse
|
45
|
Zhang Q, Song J, Wu H, Wang L, Zhuo G, Li H, He S, Pan Y, Liu G. Intratumoral microbiota associates with systemic immune inflammation state in nasopharyngeal carcinoma. Int Immunopharmacol 2024; 141:112984. [PMID: 39173404 DOI: 10.1016/j.intimp.2024.112984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/07/2024] [Accepted: 08/16/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND The nasopharynx serves as a crucial niche for the microbiome of the upper respiratory tract. However, the association between the intratumoral microbiota and host systemic inflammation and immune status in nasopharyngeal carcinoma (NPC) remain uncertain. METHODS We performed 5R 16S rDNA sequencing on NPC tissue samples, followed by diversity analysis, LEfSe differential analysis, and KEGG functional prediction. The analyses were based on indices such as AISI, SIRI, PAR, PLR, and NAR. Correlation analyses between microbes and these indices were performed to identify microbes associated with inflammation and immune status. Additionally, regression analysis based on tumor TNM stage was performed to identify key microbes linked to tumor progression. The head and neck squamous cell carcinoma (HNSC) transcriptome and the paired HNSC microbiome data from TCGA were utilized to validate the analyses. RESULTS The Proteobacteria, Actinobacteria, Firmicutes, and Bacteroidetes were the most enriched phyla in NPC tissues. Microbes within these phyla demonstrated high sensitivity to changes in host systemic inflammation and immune status. Proteobacteria and Firmicutes showed significant differences between inflammation groups. Actinobacteria varied specifically with platelet-related inflammatory indices, and Bacteroidetes genera exhibited significant differences between NAR groups. Corynebacterium and Brevundimonas significantly impacted the T stage of tumors, with a high load of Corynebacterium within tumors associated with a better prognosis CONCLUSION: Our analysis indicates that Proteobacteria play a crucial role in the inflammatory state of NPC, while Bacteroidetes are more sensitive to the tumor immune status.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China
| | - Jiangqin Song
- Department of Laboratory Medicine, The First People's Hospital of Tianmen City, Tianmen, Hubei 431700, China
| | - Huiqing Wu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China
| | - Liping Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China
| | - Guangzheng Zhuo
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China
| | - Huashun Li
- Department of Pathology, The First People's Hospital of Tianmen City, Tianmen, Hubei 431700, China
| | - Siyu He
- Department of Laboratory Medicine, The First People's Hospital of Tianmen City, Tianmen, Hubei 431700, China
| | - Yunbao Pan
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China; Hubei Engineering Center for Infectious Disease Prevention, Control and Treatment, Wuhan, China.
| | - Guohong Liu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China.
| |
Collapse
|
46
|
Yin T, Zhang X, Xiong Y, Li B, Guo D, Sha Z, Lin X, Wu H. Exploring gut microbial metabolites as key players in inhibition of cancer progression: Mechanisms and therapeutic implications. Microbiol Res 2024; 288:127871. [PMID: 39137590 DOI: 10.1016/j.micres.2024.127871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/19/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024]
Abstract
The gut microbiota plays a critical role in numerous biochemical processes essential for human health, such as metabolic regulation and immune system modulation. An increasing number of research suggests a strong association between the gut microbiota and carcinogenesis. The diverse metabolites produced by gut microbiota can modulate cellular gene expression, cell cycle dynamics, apoptosis, and immune system functions, thereby exerting a profound influence on cancer development and progression. A healthy gut microbiota promotes substance metabolism, stimulates immune responses, and thereby maintains the long-term homeostasis of the intestinal microenvironment. When the gut microbiota becomes imbalanced and disrupts the homeostasis of the intestinal microenvironment, the risk of various diseases increases. This review aims to elucidate the impact of gut microbial metabolites on cancer initiation and progression, focusing on short-chain fatty acids (SCFAs), polyamines (PAs), hydrogen sulfide (H2S), secondary bile acids (SBAs), and microbial tryptophan catabolites (MTCs). By detailing the roles and molecular mechanisms of these metabolites in cancer pathogenesis and therapy, this article sheds light on dual effects on the host at different concentrations of metabolites and offers new insights into cancer research.
Collapse
Affiliation(s)
- Tianxiang Yin
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Xiang Zhang
- Medical School, Yan'an University, Yan'an 716000, China
| | - Yan Xiong
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Bohao Li
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Dong Guo
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Zhou Sha
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Xiaoyuan Lin
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| | - Haibo Wu
- School of Life Sciences, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
47
|
Liu W, Wang X, Wu W. Role and functional mechanisms of IL‑17/IL‑17R signaling in pancreatic cancer (Review). Oncol Rep 2024; 52:144. [PMID: 39219271 PMCID: PMC11378154 DOI: 10.3892/or.2024.8803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
Interleukin‑17 (IL‑17), an inflammatory cytokine primarily secreted by T helper 17 cells, serves a crucial role in numerous inflammatory diseases and malignancies via its receptor, IL‑17R. In addition to stimulating inflammatory responses, IL‑17 exhibits dual functions in tumors, exerting both pro‑ and antitumor effects. Pancreatic ductal adenocarcinoma (PDAC) is the most common pancreatic malignancy and accounts for >90% of pancreatic cancer cases. PDAC is characterized by a prominent stromal microenvironment with significant heterogeneity, which contributes to treatment resistance. IL‑17/IL‑17R signaling has a notable effect on tumorigenesis, the tumor microenvironment and treatment efficacy in various cancer types, including PDAC. However, the specific mechanisms of IL‑17/IL‑17R signaling in pancreatic cancer remain uncertain. This review presents a brief overview of the current knowledge and recent advances in the role and functional mechanisms of IL‑17/IL‑17R signaling in pancreatic cancer. Furthermore, the potential of IL‑17‑targeted therapeutic strategies for PDAC treatment is also discussed.
Collapse
Affiliation(s)
- Wanli Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Xianze Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Wenming Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
48
|
Liu Z, Sun Y, Li Y, Ma A, Willaims NF, Jahanbahkshi S, Hoyd R, Wang X, Zhang S, Zhu J, Xu D, Spakowicz D, Ma Q, Liu B. An Explainable Graph Neural Framework to Identify Cancer-Associated Intratumoral Microbial Communities. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403393. [PMID: 39225619 PMCID: PMC11538693 DOI: 10.1002/advs.202403393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/26/2024] [Indexed: 09/04/2024]
Abstract
Microbes are extensively present among various cancer tissues and play critical roles in carcinogenesis and treatment responses. However, the underlying relationships between intratumoral microbes and tumors remain poorly understood. Here, a MIcrobial Cancer-association Analysis using a Heterogeneous graph transformer (MICAH) to identify intratumoral cancer-associated microbial communities is presented. MICAH integrates metabolic and phylogenetic relationships among microbes into a heterogeneous graph representation. It uses a graph transformer to holistically capture relationships between intratumoral microbes and cancer tissues, which improves the explainability of the associations between identified microbial communities and cancers. MICAH is applied to intratumoral bacterial data across 5 cancer types and 5 fungi datasets, and its generalizability and reproducibility are demonstrated. After experimentally testing a representative observation using a mouse model of tumor-microbe-immune interactions, a result consistent with MICAH's identified relationship is observed. Source tracking analysis reveals that the primary known contributor to a cancer-associated microbial community is the organs affected by the type of cancer. Overall, this graph neural network framework refines the number of microbes that can be used for follow-up experimental validation from thousands to tens, thereby helping to accelerate the understanding of the relationship between tumors and intratumoral microbiomes.
Collapse
Affiliation(s)
- Zhaoqian Liu
- School of MathematicsShandong UniversityJinanShandong250100China
- College of SciencesXi'an University of Science and TechnologyXi'anShanxi710054China
| | - Yuhan Sun
- School of MathematicsShandong UniversityJinanShandong250100China
| | - Yingjie Li
- Department of Biomedical InformaticsThe Ohio State UniversityColumbusOH43210USA
| | - Anjun Ma
- Department of Biomedical InformaticsThe Ohio State UniversityColumbusOH43210USA
- Pelotonia Institute for Immuno‐OncologyThe Ohio State UniversityColumbusOH43210USA
| | - Nyelia F. Willaims
- Department of Internal MedicineCollege of MedicineThe Ohio State UniversityColumbusOH43210USA
| | - Shiva Jahanbahkshi
- Department of Food Science and TechnologyCollege of FoodAgricultural, and Environmental SciencesThe Ohio State UniversityColumbusOH43210USA
| | - Rebecca Hoyd
- Department of Internal MedicineCollege of MedicineThe Ohio State UniversityColumbusOH43210USA
| | - Xiaoying Wang
- Department of Biomedical InformaticsThe Ohio State UniversityColumbusOH43210USA
- Pelotonia Institute for Immuno‐OncologyThe Ohio State UniversityColumbusOH43210USA
| | - Shiqi Zhang
- Department of Human SciencesCollege of Education and Human EcologyThe Ohio State UniversityColumbusOH43210USA
| | - Jiangjiang Zhu
- Department of Human SciencesCollege of Education and Human EcologyThe Ohio State UniversityColumbusOH43210USA
| | - Dong Xu
- Department of Electrical Engineering and Computer ScienceUniversity of MissouriColumbiaMO65201USA
- Christopher S. Bond Life Sciences CenterUniversity of MissouriColumbiaMO65201USA
| | - Daniel Spakowicz
- Pelotonia Institute for Immuno‐OncologyThe Ohio State UniversityColumbusOH43210USA
- Department of Internal MedicineCollege of MedicineThe Ohio State UniversityColumbusOH43210USA
| | - Qin Ma
- Department of Biomedical InformaticsThe Ohio State UniversityColumbusOH43210USA
- Pelotonia Institute for Immuno‐OncologyThe Ohio State UniversityColumbusOH43210USA
| | - Bingqiang Liu
- School of MathematicsShandong UniversityJinanShandong250100China
- Shandong National Center for Applied MathematicsJinanShandong250199China
| |
Collapse
|
49
|
Lombardo C, Fazio R, Sinagra M, Gattuso G, Longo F, Lombardo C, Salmeri M, Zanghì GN, Loreto CAE. Intratumoral Microbiota: Insights from Anatomical, Molecular, and Clinical Perspectives. J Pers Med 2024; 14:1083. [PMID: 39590575 PMCID: PMC11595780 DOI: 10.3390/jpm14111083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/26/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
The human microbiota represents a heterogeneous microbial community composed of several commensal, symbiotic, and even pathogenic microorganisms colonizing both the external and internal body surfaces. Despite the term "microbiota" being commonly used to identify microorganisms inhabiting the gut, several pieces of evidence suggest the presence of different microbiota physiologically colonizing other organs. In this context, several studies have also confirmed that microbes are integral components of tumor tissue in different types of cancer, constituting the so-called "intratumoral microbiota". The intratumoral microbiota is closely related to the occurrence and development of cancer as well as to the efficacy of anticancer treatments. Indeed, intratumoral microbiota can contribute to carcinogenesis and metastasis formation as some microbes can directly cause DNA damage, while others can induce the activation of proinflammatory responses or oncogenic pathways and alter the tumor microenvironment (TME). All these characteristics make the intratumoral microbiota an interesting topic to investigate for both diagnostic and prognostic purposes in order to improve the management of cancer patients. This review aims to gather the most recent data on the role of the intratumoral microbiota in cancer development, progression, and response to treatment, as well as its potential diagnostic and prognostic value.
Collapse
Affiliation(s)
- Claudia Lombardo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.L.); (R.F.); (M.S.); (C.L.); (M.S.); (C.A.E.L.)
| | - Rosanna Fazio
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.L.); (R.F.); (M.S.); (C.L.); (M.S.); (C.A.E.L.)
| | - Marta Sinagra
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.L.); (R.F.); (M.S.); (C.L.); (M.S.); (C.A.E.L.)
| | - Giuseppe Gattuso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.L.); (R.F.); (M.S.); (C.L.); (M.S.); (C.A.E.L.)
| | - Federica Longo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.L.); (R.F.); (M.S.); (C.L.); (M.S.); (C.A.E.L.)
| | - Cinzia Lombardo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.L.); (R.F.); (M.S.); (C.L.); (M.S.); (C.A.E.L.)
| | - Mario Salmeri
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.L.); (R.F.); (M.S.); (C.L.); (M.S.); (C.A.E.L.)
| | - Guido Nicola Zanghì
- Department of General Surgery and Medical-Surgical Specialties, Policlinico-Vittorio Emanuele Hospital, University of Catania, 95123 Catania, Italy;
| | - Carla Agata Erika Loreto
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.L.); (R.F.); (M.S.); (C.L.); (M.S.); (C.A.E.L.)
| |
Collapse
|
50
|
Chen G, Ren Q, Zhong Z, Li Q, Huang Z, Zhang C, Yuan H, Feng Z, Chen B, Wang N, Feng Y. Exploring the gut microbiome's role in colorectal cancer: diagnostic and prognostic implications. Front Immunol 2024; 15:1431747. [PMID: 39483461 PMCID: PMC11524876 DOI: 10.3389/fimmu.2024.1431747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
The intricate interplay between the gut microbiome and colorectal cancer (CRC) presents novel avenues for early diagnosis and prognosis, crucial for improving patient outcomes. This comprehensive review synthesizes current findings on the gut microbiome's contribution to CRC pathogenesis, highlighting its potential as a biomarker for non-invasive CRC screening strategies. We explore the mechanisms through which the microbiome influences CRC, including its roles in inflammation, metabolism, and immune response modulation. Furthermore, we assess the viability of microbial signatures as predictive tools for CRC prognosis, offering insights into personalized treatment approaches. Our analysis underscores the necessity for advanced metagenomic studies to elucidate the complex microbiome-CRC nexus, aiming to refine diagnostic accuracy and prognostic assessment in clinical settings. This review propels forward the understanding of the microbiome's diagnostic and prognostic capabilities, paving the way for microbiome-based interventions in CRC management.
Collapse
Affiliation(s)
- Guoming Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Qing Ren
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Zilan Zhong
- The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qianfan Li
- The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhiqiang Huang
- The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Hongchao Yuan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Zixin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|