1
|
Pan L, Wang X, Yang B, Liu Y, Tang D. Importance of intestinal microflora: Dried toad skin-radix clematidis plasma component analysis and anti-CRC core target study. J Pharm Biomed Anal 2025; 260:116802. [PMID: 40086049 DOI: 10.1016/j.jpba.2025.116802] [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: 09/13/2024] [Revised: 01/24/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
The focus of this study is to explore the impact of gut microbiota in different states on the blood components of couplet medications (dried toad skin and radix clematidis) and to identify drug metabolites associated with the gut microbiota. By constructing a pseudo-sterile rat model and combining non-targeted metabolomics with plasma pharmacology, we found that the plasma metabolites of couplet medications underwent significant changes in different gut microbiome environments. The GABA and PGE1 levels in the model group and the model+TCM (traditional chinese medicine) group were both significantly lower than those in the normal+TCM group. When the gut microbiota is imbalanced, drug interventions cannot significantly increase the levels of GABA and PGE1. It further confirmed the correlation between the levels of GABA and PGE1 and the gut microbiota. Based on the results of non-targeted metabolomics, we applied network pharmacology and molecular docking to explore the core targets for colorectal cancer treatment based on gut microbiota. In the end, we identified TNF and PPARG as the two core targets. These research findings provide a possibility for clarifying the molecular mechanisms of couplet medications in the treatment of colorectal cancer. It also laid the foundation for further clarifying the molecular mechanisms of Chanling Paste in the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Lijun Pan
- Department of Medical Affairs, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Xueyan Wang
- The First College of Clinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550005, China
| | - Bing Yang
- Student Management Office, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Yang Liu
- Scientific Research Section, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Dongxin Tang
- Vice President's Office, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550005, China.
| |
Collapse
|
2
|
Hamza M, Wang S, Liu Y, Li K, Zhu M, Chen L. Unraveling the potential of bioengineered microbiome-based strategies to enhance cancer immunotherapy. Microbiol Res 2025; 296:128156. [PMID: 40158322 DOI: 10.1016/j.micres.2025.128156] [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: 11/21/2024] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
The human microbiome plays a pivotal role in the field of cancer immunotherapy. The microbial communities that inhabit the gastrointestinal tract, as well as the bacterial populations within tumors, have been identified as key modulators of therapeutic outcomes, affecting immune responses and reprogramming the tumor microenvironment. Advances in synthetic biology have made it possible to reprogram and engineer these microorganisms to improve antitumor activity, enhance T-cell function, and enable targeted delivery of therapies to neoplasms. This review discusses the role of the microbiome in modulating both innate and adaptive immune mechanisms-ranging from the initiation of cytokine production and antigen presentation to the regulation of immune checkpoints-and discusses how these mechanisms improve the efficacy of immune checkpoint inhibitors. We highlight significant advances with bioengineered strains like Escherichia coli Nissle 1917, Lactococcus lactis, Bifidobacterium, and Bacteroides, which have shown promising antitumor efficacy in preclinical models. These engineered microorganisms not only efficiently colonize tumor tissues but also help overcome resistance to standard therapies by reprogramming the local immune environment. Nevertheless, several challenges remain, such as the requirement for genetic stability, effective tumor colonization, and the control of potential safety issues. In the future, the ongoing development of genetic engineering tools and the optimization of bacterial delivery systems are crucial for the translation of microbiome-based therapies into the clinic. This review highlights the potential of bioengineered microbiota as an innovative, personalized approach in cancer immunotherapy, bringing hope for more effective and personalized treatment options for patients with advanced malignancies.
Collapse
Affiliation(s)
- Muhammad Hamza
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China
| | - Yike Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Kun Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Motao Zhu
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Lin Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
3
|
Rodriguez VR, Essex M, Poddubnyy D. The gut microbiota in spondyloarthritis: an update. Curr Opin Rheumatol 2025; 37:199-206. [PMID: 39968641 DOI: 10.1097/bor.0000000000001079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
PURPOSE OF REVIEW This review provides an updated overview of the gut microbiota's involvement in spondyloarthritis (SpA) from a clinical perspective. It explores mechanisms by which the gut microbiota may influence SpA pathogenesis and considers the therapeutic implications of targeting the microbiome in SpA treatment. RECENT FINDINGS The pathogenesis of SpA is multifactorial, involving genetic predisposition, external factors and dysregulation of the immune system. Recent studies have identified alterations in the gut microbiome of patients with SpA, including changes in microbial diversity and specific taxa linked to disease activity. HLA-B27 status seems to influence gut microbiota composition, potentially impacting disease progression. In HLA-B27 transgenic rats, the association between gut microbiota and SpA development has been confirmed, supporting findings from human studies. A compromised gut barrier, influenced by proteins like zonulin, may allow microbial antigens to translocate, triggering immune responses associated with SpA. SUMMARY These findings highlight the potential for microbiota-modulating therapies, such as probiotics, prebiotics, diet and exercise, in managing SpA. However, methodological variability in human studies exposes the need for more rigorous research to better understand these associations. This may offer the opportunity to refine treatment strategies, offering a personalized approach to managing the disease.
Collapse
Affiliation(s)
- Valeria Rios Rodriguez
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Morgan Essex
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Denis Poddubnyy
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Division of Rheumatology, University of Toronto and University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Arp G, Levy S, Hall B. Linking bacterial androgen production and prostate cancer. Nat Microbiol 2025:10.1038/s41564-025-01996-8. [PMID: 40259018 DOI: 10.1038/s41564-025-01996-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Affiliation(s)
- Gabriela Arp
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, College Park, MD, USA
| | - Sophia Levy
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, College Park, MD, USA
| | - Brantley Hall
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, College Park, MD, USA.
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, College Park, MD, USA.
| |
Collapse
|
5
|
Ren J, Yan G, Yang L, Kong L, Guan Y, Sun H, Liu C, Liu L, Han Y, Wang X. Cancer chemoprevention: signaling pathways and strategic approaches. Signal Transduct Target Ther 2025; 10:113. [PMID: 40246868 PMCID: PMC12006474 DOI: 10.1038/s41392-025-02167-1] [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: 08/02/2024] [Revised: 12/01/2024] [Accepted: 02/04/2025] [Indexed: 04/19/2025] Open
Abstract
Although cancer chemopreventive agents have been confirmed to effectively protect high-risk populations from cancer invasion or recurrence, only over ten drugs have been approved by the U.S. Food and Drug Administration. Therefore, screening potent cancer chemopreventive agents is crucial to reduce the constantly increasing incidence and mortality rate of cancer. Considering the lengthy prevention process, an ideal chemopreventive agent should be nontoxic, inexpensive, and oral. Natural compounds have become a natural treasure reservoir for cancer chemoprevention because of their superior ease of availability, cost-effectiveness, and safety. The benefits of natural compounds as chemopreventive agents in cancer prevention have been confirmed in various studies. In light of this, the present review is intended to fully delineate the entire scope of cancer chemoprevention, and primarily focuses on various aspects of cancer chemoprevention based on natural compounds, specifically focusing on the mechanism of action of natural compounds in cancer prevention, and discussing in detail how they exert cancer prevention effects by affecting classical signaling pathways, immune checkpoints, and gut microbiome. We also introduce novel cancer chemoprevention strategies and summarize the role of natural compounds in improving chemotherapy regimens. Furthermore, we describe strategies for discovering anticancer compounds with low abundance and high activity, revealing the broad prospects of natural compounds in drug discovery for cancer chemoprevention. Moreover, we associate cancer chemoprevention with precision medicine, and discuss the challenges encountered in cancer chemoprevention. Finally, we emphasize the transformative potential of natural compounds in advancing the field of cancer chemoprevention and their ability to introduce more effective and less toxic preventive options for oncology.
Collapse
Affiliation(s)
- Junling Ren
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Guangli Yan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Le Yang
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China
| | - Ling Kong
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Yu Guan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Hui Sun
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
| | - Chang Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Lei Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Ying Han
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Xijun Wang
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China.
| |
Collapse
|
6
|
Hog L, Fundin BT, Everett Palm E, Billger A, Bulik CM, Abbaspour A, Dinkler L. ARFID InitiativE Sweden (ARIES): study protocol for a large-scale genetic and registry-linked cohort study on avoidant/restrictive food intake disorder. BMJ Open 2025; 15:e095559. [PMID: 40246566 PMCID: PMC12007039 DOI: 10.1136/bmjopen-2024-095559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/19/2025] [Indexed: 04/19/2025] Open
Abstract
INTRODUCTION The ARFID InitiativE Sweden (ARIES) investigates the genetic and environmental factors contributing to avoidant/restrictive food intake disorder (ARFID) in children and adolescents aged 6-14 years. ARIES will establish a national biobank and research registry. It aims to provide data for immediate research and track ARFID outcomes and clarify genetic links between ARFID and other conditions and analyse the gut microbiome to guide nutrition interventions. METHODS AND ANALYSIS The study will involve 1500 Swedish children and adolescents with ARFID and a control group of 500 Swedish children and adolescents without ARFID. Parents/guardians and their children will complete online questionnaires assessing ARFID and other eating disorder (ED) pathology, co-occurring conditions, quality of life and parental stress and ED pathology. All participants will provide a saliva sample for comprehensive genetic analyses. Additionally, a subset of participants will provide a stool sample to investigate the gut microbiome in ARFID. ETHICS AND DISSEMINATION ARIES was approved by the Swedish Ethical Review Authority (Dnr 2023-04638). All participants will give assent and their parents will complete informed consent. Data will be made available by the authors on reasonable request. Findings will be published in scientific journals and shared with the public and stakeholders in accessible ways, for example, via social media.
Collapse
Affiliation(s)
- Liv Hog
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Bengt T Fundin
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Erik Everett Palm
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Annelie Billger
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Cynthia M Bulik
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
- Department of Psychiatry, The University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Nutrition, The University of North Carolina, Chapel Hill, North Carolina, USA
| | - Afrouz Abbaspour
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Lisa Dinkler
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
7
|
Fan Z, Yi Z, Li S, He J. Parabacteroides distasonis promotes CXCL9 secretion of tumor-associated macrophages and enhances CD8 +T cell activity to trigger anti-tumor immunity against anti-PD-1 treatment in non-small cell lung cancer mice. BMC Biotechnol 2025; 25:30. [PMID: 40241108 PMCID: PMC12004837 DOI: 10.1186/s12896-025-00963-9] [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: 09/26/2024] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Parabacteroides distasonis (P. distasonis) could regulate inflammatory markers, promote intestinal barrier integrity, and block tumor formation in colon. However, the regulatory effect of P. distasonis on non-small cell lung cancer (NSCLC) remains unknown. This study aimed to investigate the regulatory effect of P. distasonis on NSCLC and its impact on tumor immunity. METHODS We first established a mouse model of Lewis lung cancer, and administered P. distasonis and intrabitoneal injection of anti-mouse PD-1 monoclonal antibody to assess the impact of P. distasonis on tumor immunity, and mouse intestinal barrier. Then, we explored the effect of P. distasonis on CD8+T cells and CXCL9 secretion mediated by tumor-associated macrophages (TAM). We used the TLR1/2 complex inhibitor CPT22 to evaluate its effect on macrophage activation. Finally, we explored the effect of P. distasonis on CD8+T cells and CXCL9 secreted by TAM in vivo. RESULTS In vivo, P. distasonis enhanced anti-tumor effects of anti-PD-1 in NSCLC mice, improved intestinal barrier integrity, recruited macrophages, and promoted M1 polarization. In vitro, CD86 and iNOS levels in BMDM were elevated and CD206 and Arg1 levels were suppressed in membrane fraction of P. distasonis (PdMb) group in comparison to Control group. With additional CPT22 pre-treatment, the levels of CD86 and iNOS in BMDM were reduced, and the levels of CD206 and Arg1 were increased. Compared to PBS group, P. distasonis group exhibited higher proportion of CD8+T cells in tumor tissues, along with increased positive proportion of GZMB and IFN-γ in CD8+T cells. Additionally, in comparison to Control group, PdMb group showed an elevated proportion of GZMB+T and IFN-γ+T cells within CD8+T cells, and secretion of IFN-γ, TNF-α, perforin, and GZMB in CD8+T cell supernatant increased. Moreover, the proportion of CXCL9+F4/80+ macrophages in tumor tissues was higher in P. distasonis group compared to PBS group. In comparison to Control group, CXCL9 protein level in BMDM and CXCL9 secretion level in BMDM supernatant were increased in PdMb group. Finally, P. distasonis enhanced CD8+T cell activity by secreting CXCL9 from macrophages in vivo. CONCLUSIONS P. distasonis promoted CXCL9 secretion of TAM and enhanced CD8+T cell activity to trigger anti-tumor immunity against anti-PD-1 treatment in NSCLC mice.
Collapse
Affiliation(s)
- Zhijun Fan
- Department of Cardiothoracic Surgery, The People's Hospital of Liuyang, Changsha, China
| | - Zheng Yi
- Department of Cardiothoracic Surgery, The People's Hospital of Liuyang, Changsha, China
| | - Sheng Li
- Department of Gastrointestinal Surgery, The Central Hospital of Shaoyang, Shaoyang, China
| | - Junjun He
- Department of Gastrointestinal Surgery, The Central Hospital of Shaoyang, Shaoyang, China.
| |
Collapse
|
8
|
Choi SW, Kim JH, Hong J, Kwon M. Mapping immunotherapy potential: spatial transcriptomics in the unraveling of tumor-immune microenvironments in head and neck squamous cell carcinoma. Front Immunol 2025; 16:1568590. [PMID: 40264779 PMCID: PMC12011851 DOI: 10.3389/fimmu.2025.1568590] [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: 01/30/2025] [Accepted: 03/25/2025] [Indexed: 04/24/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) often exhibits poor response rates to immune checkpoint inhibitor (ICI) therapies, largely owing to the intricate composition and spatial organization of immune cells within the tumor-immune microenvironment (TIME). The diversity of immune cell populations, their spatial relationships, and dynamic interactions significantly influence the immunosuppressive nature of the TIME, thereby limiting the efficacy of immunotherapy. To address these challenges and enhance the therapeutic potential of ICIs in HNSCC, a comprehensive analysis of the TIME is essential. Spatial transcriptomics (ST), a cutting-edge technology, enables high-resolution mapping of gene expression within the spatial context of the tumor, providing critical insights into the functional roles and interactions of immune cells in the TIME. This review highlights the importance of ST in uncovering the complexities of the TIME in HNSCC and proposes strategies for leveraging these insights to develop more effective immunotherapeutic approaches. By integrating spatial and molecular information, this review aims to pave the way for personalized and precision-based treatments in HNSCC, ultimately improving patient outcomes.
Collapse
Affiliation(s)
| | | | | | - Minsu Kwon
- Department of Otolaryngology-Head and Neck Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
9
|
Fryer E, Hatcher C, Knight R, Wade KH. Exploring the causal role of the human gut microbiome in endometrial cancer: a Mendelian randomization approach. Sci Rep 2025; 15:11953. [PMID: 40199948 PMCID: PMC11978776 DOI: 10.1038/s41598-025-96740-x] [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: 11/12/2024] [Accepted: 03/31/2025] [Indexed: 04/10/2025] Open
Abstract
Endometrial cancer presents a major public health issue, particularly in post-menopausal women. Whilst there are known risk factors for the disease, including oestrogen and obesity, these factors do not fully explain risk variability in cancer outcomes. The identification of novel risk factors may aid in better understanding of endometrial cancer development and, given the link with oestrogen metabolism, obesity and the risk of various cancers, the gut microbiome could be one such risk factor. Mendelian randomization (MR), a method that reduces biases of conventional epidemiological studies (namely, confounding and reverse causation) by using genetic variants to proxy exposures, was used to investigate the effect of gut microbial traits on endometrial cancer risk. Whilst our initial analyses showed that the presence of an unclassified group of bacteria in the Erysipelotrichaceae family increased the risk of oestrogen-dependent endometrial cancer (odds ratio (OR) per approximate doubling of the genetic liability to presence vs. absence: 1.13; 95% CI 1.01, 1.26; P = 0.03), subsequent sensitivity analyses, including colocalisation, provided insufficient evidence to support causality. This work highlights the importance of using a robust MR analysis pipeline, including sensitivity analyses to assess the validity of causal effect estimates obtained using MR.
Collapse
Affiliation(s)
- Ella Fryer
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK.
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK.
| | - Charlie Hatcher
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Rochelle Knight
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Kaitlin H Wade
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| |
Collapse
|
10
|
Rahim MA, Seo H, Barman I, Hossain MS, Shuvo MSH, Song HY. Insights into Autophagy in Microbiome Therapeutic Approaches for Drug-Resistant Tuberculosis. Cells 2025; 14:540. [PMID: 40214493 PMCID: PMC11989032 DOI: 10.3390/cells14070540] [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: 02/21/2025] [Revised: 03/23/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Tuberculosis, primarily caused by Mycobacterium tuberculosis, is an airborne lung disease and continues to pose a significant global health threat, resulting in millions of deaths annually. The current treatment for tuberculosis involves a prolonged regimen of antibiotics, which leads to complications such as recurrence, drug resistance, reinfection, and a range of side effects. This scenario underscores the urgent need for novel therapeutic strategies to combat this lethal pathogen. Over the last two decades, microbiome therapeutics have emerged as promising next-generation drug candidates, offering advantages over traditional medications. In 2022, the Food and Drug Administration approved the first microbiome therapeutic for recurrent Clostridium infections, and extensive research is underway on microbiome treatments for various challenging diseases, including metabolic disorders and cancer. Research on microbiomes concerning tuberculosis commenced roughly a decade ago, and the scope of this research has broadened considerably over the last five years, with microbiome therapeutics now viewed as viable options for managing drug-resistant tuberculosis. Nevertheless, the understanding of their mechanisms is still in its infancy. Although autophagy has been extensively studied in other diseases, research into its role in tuberculosis is just beginning, with preliminary developments in progress. Against this backdrop, this comprehensive review begins by succinctly outlining tuberculosis' characteristics and assessing existing treatments' strengths and weaknesses, followed by a detailed examination of microbiome-based therapeutic approaches for drug-resistant tuberculosis. Additionally, this review focuses on establishing a basic understanding of microbiome treatments for tuberculosis, mainly through the lens of autophagy as a mechanism of action. Ultimately, this review aims to contribute to the foundational comprehension of microbiome-based therapies for tuberculosis, thereby setting the stage for the further advancement of microbiome therapeutics for drug-resistant tuberculosis.
Collapse
Affiliation(s)
- Md Abdur Rahim
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Hoonhee Seo
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
- Probiotics Microbiome Commercialization Research Center (PMC), Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Indrajeet Barman
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Mohammed Solayman Hossain
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Md Sarower Hossen Shuvo
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Ho-Yeon Song
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
- Probiotics Microbiome Commercialization Research Center (PMC), Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| |
Collapse
|
11
|
Liu CG, Lin MX, Xin Y, Sun M, Cui J, Liu D, Zang D, Chen J. Metagenomics and Non-Targeted Metabolomics Reveal the Role of Gut Microbiota and Its Metabolites in Brain Metastasis of Non-Small Cell Lung Cancer. Thorac Cancer 2025; 16:e70068. [PMID: 40263747 DOI: 10.1111/1759-7714.70068] [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: 02/15/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Brain metastasis is a common and severe complication in non-small cell lung cancer (NSCLC) patients, significantly affecting prognosis. However, the role of gut microbiota and its metabolites in NSCLC brain metastasis remains poorly understood. This study aims to explore the relationship between gut microbiota, metabolites, and the development of brain metastasis in NSCLC. METHODS We conducted an integrative analysis combining metagenomics and non-targeted metabolomics on baseline fecal samples from NSCLC patients with brain metastasis (n = 18) and those without distant metastasis (n = 12). Gut microbiota composition and metabolite profiles were detected and analyzed, and statistical methods, including machine learning models, were applied to identify differences and potential biomarkers. RESULTS Significant differences in gut microbiota composition were found between the two groups, with higher microbial diversity observed in patients with brain metastasis. Specific genera, such as Paenibacillus, Fournierella, and Adlercreutzia, were enriched in the brain metastasis group. Metabolomic analysis revealed altered levels of short-chain fatty acids and other metabolites associated with immune modulation and vascular permeability, including angiotensin (1-7). These changes were linked to the metastatic process and may influence brain metastasis development. Furthermore, machine learning models identified key biomarkers, such as Raoultibacter, Mobilibacterium, and N-acetyl-L-glutamic acid, which could serve as valuable indicators for brain metastasis. CONCLUSIONS Our findings suggest that gut microbiota dysbiosis and its metabolic products may contribute to the development of brain metastasis in NSCLC. The identification of microbiota-derived biomarkers holds potential for early detection and therapeutic intervention in NSCLC brain metastasis.
Collapse
Affiliation(s)
- Chen-Guang Liu
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Mei-Xi Lin
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Yu Xin
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Man Sun
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Jia Cui
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Dan Liu
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Dan Zang
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Jun Chen
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
12
|
Rao UK, Majhail NS, Blunk B, Abernathy K, Bachier C, Bhushan V, Cruz JC, Elayan M, Gregory T, LeMaistre CF, Malik SA, Martin C, Mattlin M, Blade G, Maris MB, Mathews J, Mountjoy L, Pantin JM, Ramakrishnan A, Shaughnessy P, Tees MT, Vance EA, Zoghi B, Battiwalla M. Comparative Efficacy of Bendamustine Versus Fludarabine/Cyclophosphamide for Lymphodepletion Before Chimeric Antigen Receptor T-Cell Therapy in Lymphoma. Transplant Cell Ther 2025:S2666-6367(25)01101-7. [PMID: 40122282 DOI: 10.1016/j.jtct.2025.03.012] [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: 09/23/2024] [Revised: 01/13/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Chimeric antigen receptor T-cell (CAR-T) therapy represents a transformative advance in treating relapsed/refractory non-Hodgkin lymphomas (NHLs). Effective pre-infusion lymphodepleting chemotherapy (LDC) is essential for optimizing CAR-T outcomes. Traditionally, a combination of fludarabine and cyclophosphamide (Flu/Cy) have been used; however, a global fludarabine shortage has necessitated alternative regimens. This study compares the efficacy and safety of bendamustine versus Flu/Cy as LDC in NHL patients. OBJECTIVES The purpose of this study was to compare the efficacy and safety of bendamustine versus Flu/Cy as LDC regimens in patients with relapsed/refractory NHL undergoing CAR-T therapy. We hypothesized that bendamustine would offer comparable outcomes to Flu/Cy while providing logistical advantages in outpatient settings. STUDY DESIGN This retrospective analysis evaluated 265 NHL patients treated with FDA-approved CAR-T products from January 2018 to October 2023. Outcomes included cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), hematologic recovery, progression-free survival (PFS), overall survival (OS), and healthcare resource utilization. Kaplan-Meier survival analysis and multivariable Cox proportional hazards models were employed to adjust for CAR-T product type, infusion year, disease subtype, and dexamethasone prophylaxis. RESULTS Both LDC regimens effectively prepared patients for CAR-T therapy, with no significant differences in CRS, ICANS, OS, or PFS. At one year, OS was 71% for bendamustine versus 68% for Flu/Cy, and PFS was 68% versus 60% (P = .3 and P = .4, respectively). Bendamustine was associated with less severe hematologic toxicity; ANC levels did not fall below 0.5 K/µL in 71% of bendamustine recipients compared to 17% for Flu/Cy (P < .001). Multivariable analysis identified infusion year as a significant predictor of OS (HR 0.77, P = .008) and PFS (HR 2.6, P < .001), reflecting improvements in CAR-T practices over time. CONCLUSION Bendamustine is a viable alternative to Flu/Cy for LDC prior to CAR-T therapy in relapsed/refractory NHL, as it demonstrates comparable efficacy and safety. Its operational advantages, including reduced hospitalization rates and suitability for outpatient administration, underscore its potential in diverse clinical settings. Prospective studies are needed to confirm long-term outcomes and further optimize LDC strategies.
Collapse
Affiliation(s)
- Uttam K Rao
- Sarah Cannon Transplant and Cellular Therapy Program at St. David's South Austin Medical Center, Austin, Texas.
| | - Navneet S Majhail
- Sarah Cannon Transplant and Cellular Therapy Network, Nashville, Tennessee; Sarah Cannon Transplant and Cellular Therapy Program at TriStar Centennial Medical Center, Nashville, Tennessee
| | - Betsy Blunk
- Sarah Cannon Transplant and Cellular Therapy Network, Nashville, Tennessee
| | - Karin Abernathy
- Sarah Cannon Transplant and Cellular Therapy Program at TriStar Centennial Medical Center, Nashville, Tennessee
| | - Carlos Bachier
- Sarah Cannon Transplant and Cellular Therapy Program at Methodist Hospital, San Antonio, Texas
| | - Vikas Bhushan
- Sarah Cannon and Texas Oncology Transplant and Cellular Therapy Program at Medical City Dallas, Dallas, Texas
| | - Jose Carlos Cruz
- Sarah Cannon Transplant and Cellular Therapy Program at Methodist Hospital, San Antonio, Texas
| | - Mohammed Elayan
- Sarah Cannon Transplant and Cellular Therapy Network, Nashville, Tennessee
| | - Tara Gregory
- Sarah Cannon Transplant and Cellular Therapy Program at Presbyterian/St Luke's Medical Center, Denver, Colorado; Colorado Blood Cancer Institute, Denver, Colorado
| | | | - Shahbaz A Malik
- Sarah Cannon Transplant and Cellular Therapy Program at St. David's South Austin Medical Center, Austin, Texas
| | | | | | | | - Michael B Maris
- Sarah Cannon Transplant and Cellular Therapy Program at Presbyterian/St Luke's Medical Center, Denver, Colorado; Colorado Blood Cancer Institute, Denver, Colorado
| | - John Mathews
- Sarah Cannon and Texas Oncology Transplant and Cellular Therapy Program at Medical City Dallas, Dallas, Texas
| | - Luke Mountjoy
- Sarah Cannon Transplant and Cellular Therapy Program at Presbyterian/St Luke's Medical Center, Denver, Colorado; Colorado Blood Cancer Institute, Denver, Colorado
| | - Jeremy M Pantin
- Sarah Cannon Transplant and Cellular Therapy Program at TriStar Centennial Medical Center, Nashville, Tennessee
| | - Aravind Ramakrishnan
- Sarah Cannon Transplant and Cellular Therapy Program at St. David's South Austin Medical Center, Austin, Texas
| | - Paul Shaughnessy
- Sarah Cannon Transplant and Cellular Therapy Program at Methodist Hospital, San Antonio, Texas
| | - Michael T Tees
- Sarah Cannon Transplant and Cellular Therapy Program at Presbyterian/St Luke's Medical Center, Denver, Colorado; Colorado Blood Cancer Institute, Denver, Colorado
| | - Estil A Vance
- Sarah Cannon and Texas Oncology Transplant and Cellular Therapy Program at Medical City Dallas, Dallas, Texas
| | - Behyar Zoghi
- Sarah Cannon Transplant and Cellular Therapy Program at Methodist Hospital, San Antonio, Texas
| | - Minoo Battiwalla
- Sarah Cannon Transplant and Cellular Therapy Network, Nashville, Tennessee; Sarah Cannon Transplant and Cellular Therapy Program at TriStar Centennial Medical Center, Nashville, Tennessee
| |
Collapse
|
13
|
Sharma P, Jain T, Sorgen A, Iyer S, Tarique M, Roy P, Kurtom S, Sethi V, Bava EP, Gutierrez-Garcia AK, Vaish U, Suresh DS, Sahay P, Edwards D, Afghani J, Putluri S, Reddy KRK, Amara CS, Kamal AHM, Fodor A, Dudeja V. Smoking-induced gut microbial dysbiosis mediates cancer progression through modulation of anti-tumor immune response. iScience 2025; 28:112002. [PMID: 40104059 PMCID: PMC11914281 DOI: 10.1016/j.isci.2025.112002] [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: 05/01/2024] [Revised: 11/01/2024] [Accepted: 02/07/2025] [Indexed: 03/20/2025] Open
Abstract
Cigarette smoke exposure (CSE) increases the risk for a plethora of cancers. Recent evidence indicates that the gut microbiome can influence cancer progression by immune system modulation. Since CSE alters the gut microbiome, we hypothesized that the gut microbiome serves as a causative link between smoking and cancer growth. Through a combination of syngeneic animal models and fecal microbiota transplantation studies, we established an essential role for smoke-induced dysbiosis in cancer growth. 16s rRNA sequencing and liquid chromatography-mass spectrometry indicated a unique CSE-associated microbial and metabolomic signature. Immunophenotyping of tumor specimens and experiments in Rag1-KO and CD8-KO demonstrated that smoke-induced tumor growth requires functional adaptive immunity. Finally, utilizing gut microbial ablation strategies with broad- and narrow-spectrum antibiotics, we demonstrated the reversal of phenotypic effects of CSE. Our study provides evidence for gut microbiome as an actionable target to mitigate CSE-induced tumor promotion.
Collapse
Affiliation(s)
- Prateek Sharma
- Department of Surgery, University of Alabama, Birmingham, AL 35233, USA
| | - Tejeshwar Jain
- Department of Surgery, University of Alabama, Birmingham, AL 35233, USA
| | - Ali Sorgen
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Srikanth Iyer
- Department of Surgery, University of Alabama, Birmingham, AL 35233, USA
| | - Mohammad Tarique
- Sylvester Cancer Center, Department of Surgery, University of Miami, Miami, FL 33136, USA
| | - Pooja Roy
- Sylvester Cancer Center, Department of Surgery, University of Miami, Miami, FL 33136, USA
| | - Saba Kurtom
- Sylvester Cancer Center, Department of Surgery, University of Miami, Miami, FL 33136, USA
| | - Vrishketan Sethi
- Department of Surgery, University of Alabama, Birmingham, AL 35233, USA
| | - Ejas P Bava
- Department of Surgery, University of Alabama, Birmingham, AL 35233, USA
| | | | - Utpreksha Vaish
- Department of Surgery, University of Alabama, Birmingham, AL 35233, USA
| | | | - Preeti Sahay
- Department of Surgery, University of Alabama, Birmingham, AL 35233, USA
| | - Dujon Edwards
- Sylvester Cancer Center, Department of Surgery, University of Miami, Miami, FL 33136, USA
| | - Jumana Afghani
- Sylvester Cancer Center, Department of Surgery, University of Miami, Miami, FL 33136, USA
| | - Satwikreddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Karthik Reddy Kami Reddy
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chandra Sekhar Amara
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Abu Hena Mustafa Kamal
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Anthony Fodor
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Vikas Dudeja
- Department of Surgery, University of Alabama, Birmingham, AL 35233, USA
| |
Collapse
|
14
|
Qin Y, Wang Q, Lin Q, Liu F, Pan X, Wei C, Chen J, Huang T, Fang M, Yang W, Pan L. Multi-omics analysis reveals associations between gut microbiota and host transcriptome in colon cancer patients. mSystems 2025; 10:e0080524. [PMID: 40013792 PMCID: PMC11915798 DOI: 10.1128/msystems.00805-24] [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: 06/13/2024] [Accepted: 01/31/2025] [Indexed: 02/28/2025] Open
Abstract
Colon cancer (CC) is one of the most common cancers globally, which is associated with the gut microbiota intimately. In current research, exploring the complex interaction between microbiomes and CC is a hotspot. However, the information on microbiomes in most previous studies is based on fecal, which does not fully display the microbial environment of CC. Herein, we collected mucosal and tissue samples from both the tumor and normal regions of 19 CC patients and clarified the composition of mucosal microbiota by 16S rRNA and metagenomic sequencing. Additionally, RNA-Seq was also conducted to identify the different expression genes between tumor and normal tissue samples. We revealed significantly different microbial community structures and expression profiles to CC. Depending on correlation analysis, we demonstrated that 1,472 genes were significantly correlated with CC tumor microbiota. Our study reveals a significant enrichment of Campylobacter jejuni in the mucosa of CC, which correlates with bile secretion. Additionally, we observe a negative correlation between C. jejuni and immune cells CD4+ Tem and mast cells. Finally, we discovered that metabolic bacterial endosymbiont of Bathymodiolus sp., Bacillus wiedmannii, and Mycobacterium tuberculosis had a significant survival value for CC, which was ignored by previous research. Overall, our study expands the understanding of the complex interplay between microbiota and CC and provides new targets for the treatment of CC. IMPORTANCE This study contributes to our understanding of the interaction between microbiota and colon cancer (CC). By examining mucosal and tissue samples rather than solely relying on fecal samples, we have uncovered previously unknown aspects of CC-associated microbiota. Our findings reveal distinct microbial community structures and gene expression profiles correlated with CC progression. Notably, the enrichment of Campylobacter jejuni in CC mucosa, linked to bile secretion, underscores potential mechanisms in CC pathogenesis. Additionally, observed correlations between microbial taxa and immune cell populations offer new avenues for immunotherapy research in CC. Importantly, this study introduces CC-associated microbiota with survival implications for CC, expanding therapeutic targets beyond conventional strategies. By elucidating these correlations, our study not only contributes to uncovering the potential role of gut microbiota in colon cancer but also establishes a foundation for mechanistic studies of gut microbiota in colon cancer, emphasizing the broader impact of microbiota research on cancer biology.
Collapse
Affiliation(s)
- Yuling Qin
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qiang Wang
- Guangxi Clinical Research Center for Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qiumei Lin
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Fengfei Liu
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiaolan Pan
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Caibiao Wei
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Junxian Chen
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Taijun Huang
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Min Fang
- Guangxi Clinical Research Center for Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Weilong Yang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Institute of Advanced Biotechnology and School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Linghui Pan
- Guangxi Clinical Research Center for Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
15
|
Schieferecke AJ, Kuxhausen Ralph N, Schaffer DV. The Application of DNA Viruses to Biotechnology. Viruses 2025; 17:414. [PMID: 40143341 PMCID: PMC11946468 DOI: 10.3390/v17030414] [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: 02/01/2025] [Revised: 02/24/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
The delivery of biomolecules to target cells has been a longstanding challenge in biotechnology. DNA viruses naturally evolved the ability to deliver genetic material to cells and modulate cellular processes. As such, they inherently possess requisite characteristics that have led to their extensive study, engineering, and development as biotechnological tools. Here, we overview the application of DNA viruses to biotechnology, with specific implications in basic research, health, biomanufacturing, and agriculture. For each application, we review how an increasing understanding of virology and technological methods to genetically manipulate DNA viruses has enabled advances in these fields. Additionally, we highlight the remaining challenges to unlocking the full biotechnological potential of DNA viral technologies. Finally, we discuss the importance of balancing continued technological progress with ethical and biosafety considerations.
Collapse
Affiliation(s)
- Adam J. Schieferecke
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; (N.K.R.); (D.V.S.)
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Nadia Kuxhausen Ralph
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; (N.K.R.); (D.V.S.)
| | - David V. Schaffer
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; (N.K.R.); (D.V.S.)
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
16
|
Zhang X, Chen Y, Liu X, Li G, Zhang S, Zhang Q, Cui Z, Qin M, Simon HU, Terzić J, Kocic G, Polić B, Yin C, Li X, Zheng T, Liu B, Zhu Y. STING in cancer immunoediting: Modeling tumor-immune dynamics throughout cancer development. Cancer Lett 2025; 612:217410. [PMID: 39826670 DOI: 10.1016/j.canlet.2024.217410] [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: 09/09/2024] [Revised: 12/16/2024] [Accepted: 12/21/2024] [Indexed: 01/22/2025]
Abstract
Cancer immunoediting is a dynamic process of tumor-immune system interaction that plays a critical role in cancer development and progression. Recent studies have highlighted the importance of innate signaling pathways possessed by both cancer cells and immune cells in this process. The STING molecule, a pivotal innate immune signaling molecule, mediates DNA-triggered immune responses in both cancer cells and immune cells, modulating the anti-tumor immune response and shaping the efficacy of immunotherapy. Emerging evidence has shown that the activation of STING signaling has dual opposing effects in cancer progression, simultaneously provoking and restricting anti-tumor immunity, and participating in every phase of cancer immunoediting, including immune elimination, equilibrium, and escape. In this review, we elucidate the roles of STING in the process of cancer immunoediting and discuss the dichotomous effects of STING agonists in the cancer immunotherapy response or resistance. A profound understanding of the sophisticated roles of STING signaling pathway in cancer immunoediting would potentially inspire the development of novel cancer therapeutic approaches and overcome the undesirable protumor effects of STING activation.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China; Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Yan Chen
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Xi Liu
- Department of Cardiology, ordos central hospital, Ordos, People's Republic of China
| | - Guoli Li
- Department of Colorectal and Anal Surgery, Chifeng Municipal Hospital, Chifeng Clinical Medical School of Inner Mongolia Medical University, Chifeng, People's Republic of China
| | - Shuo Zhang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China
| | - Qi Zhang
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Zihan Cui
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Minglu Qin
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland; Institute of Biochemistry, Brandenburg Medical School, Neuruppin, 16816, Germany
| | - Janoš Terzić
- Laboratory for Cancer Research, University of Split School of Medicine, Split, Croatia
| | - Gordana Kocic
- Department of Biochemistry, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
| | - Bojan Polić
- University of Rijeka Faculty of Medicine, Croatia
| | - Chengliang Yin
- Faculty of Medicine, Macau University of Science and Technology, 999078, Macao.
| | - Xiaobo Li
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China.
| | - Tongsen Zheng
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No.150 Haping Road, Nangang District, Harbin, Heilongjiang, People's Republic of China.
| | - Bing Liu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China; School of Stomatology, Harbin Medical University, Harbin, 150001, People's Republic of China.
| | - Yuanyuan Zhu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China; Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China.
| |
Collapse
|
17
|
Li C, Faiz SA, Boysen-Osborn M, Sheshadri A, Wattana MK. Immune Checkpoint Inhibitor-associated Pneumonitis: A Narrative Review. West J Emerg Med 2025; 26:210-218. [PMID: 40145913 PMCID: PMC11931710 DOI: 10.5811/westjem.20305] [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: 03/04/2024] [Revised: 09/12/2024] [Accepted: 12/07/2024] [Indexed: 03/28/2025] Open
Abstract
Immune checkpoint inhibitors (ICI), such as pembrolizumab, nivolumab, durvalumab and ipilimumab, have significantly enhanced survival rates for multiple cancer types such as non-small cell lung cancer, melanoma, Hodgkin lymphoma, and breast cancer, and they have emerged as an adjunct or primary therapy for malignant disease. Approximately 40% of patients with cancer on ICI therapy experience side effects called immune-related adverse events (irAE). While not the most common, pulmonary toxicities can be rapidly progressive, potentially fatal, and pose a three-fold increased risk for requiring intensive care unit-level of care. Pneumonitis is a focal or diffuse inflammation of the lung parenchyma, and clinical manifestations may be highly variable. While the onset is generally observed 6-12 weeks after the initiation of therapy, drug toxicity can develop rapidly within days after the first infusion or many months into therapy. Pneumonitis symptoms can be subtle or non-specific; therefore, a thorough and systematic evaluation considering other possible etiologies is crucial. Moreover, extrapulmonary findings, such as skin lesions, colitis, or endocrinopathies, should raise suspicion for irAE as drug toxicity can affect multiple organs simultaneously. Due to the significant overlap of clinical features between ICI-associated pneumonitis and respiratory infections, it can be challenging to differentiate the two conditions based on clinical presentation alone. A multidisciplinary approach to management is recommended for the treatment of ICI-associated pneumonitis, and classification of severity helps to guide interventions. Treatment options in more severe cases include systemic immunosuppression. Given the increased use of ICIs and greater probability that patients with ICI-associated pneumonitis will be seen in the emergency department, we aimed to provide a comprehensive framework for the diagnosis and management. In addition, identifying potential challenges in diagnosis and/or other contributors of respiratory symptoms and radiographic manifestations is highlighted.
Collapse
Affiliation(s)
- Chang Li
- McGovern Medical School at University of Texas Health, Divisions of Pulmonary, Critical Care Medicine and Sleep Medicine, Houston, Texas
| | - Saadia A Faiz
- The University of Texas MD Anderson Cancer Center, Department of Pulmonary Medicine, Houston, Texas
| | - Megan Boysen-Osborn
- University of California Irvine School of Medicine, Department of Emergency Medicine, Irvine, California
| | - Ajay Sheshadri
- The University of Texas MD Anderson Cancer Center, Department of Pulmonary Medicine, Houston, Texas
| | - Monica K Wattana
- The University of Texas MD Anderson Cancer Center, Department of Emergency Medicine, Houston, Texas
| |
Collapse
|
18
|
Chacon J, Faizuddin F, McKee JC, Sheikh A, Vasquez VM, Gadad SS, Mayer G, Siby S, McCabe M, Dhandayuthapani S. Unlocking the Microbial Symphony: The Interplay of Human Microbiota in Cancer Immunotherapy Response. Cancers (Basel) 2025; 17:813. [PMID: 40075661 PMCID: PMC11899421 DOI: 10.3390/cancers17050813] [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: 01/11/2025] [Revised: 02/13/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
INTRODUCTION The emergence of cancer immunotherapy has revolutionized cancer treatment, offering remarkable outcomes for patients across various malignancies. However, the heterogeneous response to immunotherapy underscores the necessity of understanding additional factors influencing treatment efficacy. Among these factors, the human microbiota has garnered significant attention for its potential role in modulating immune response. Body: This review explores the intricate relationship between the human microbiota and cancer immunotherapy, highlighting recent advances and potential mechanisms underlying microbial influence on treatment outcomes. CONCLUSION Insights into the microbiome's impact on immunotherapy response not only deepen our understanding of cancer pathogenesis but also hold promise for personalized therapeutic strategies aimed at optimizing patient outcomes.
Collapse
Affiliation(s)
- Jessica Chacon
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
| | - Farah Faizuddin
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
| | - Jack C. McKee
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
| | - Aadil Sheikh
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
| | - Victor M. Vasquez
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
| | - Shrikanth S. Gadad
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
- L. Frederick Francis Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Ghislaine Mayer
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
| | - Sharon Siby
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
| | - Molly McCabe
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
| | - Subramanian Dhandayuthapani
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
- L. Frederick Francis Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| |
Collapse
|
19
|
Braun A, Deng M, Hasler JS, Bukavina L, Handorf E, Abbosh PH. Association between antibiotics and treatment efficacy in metastatic urothelial carcinoma patients. BMC Med 2025; 23:117. [PMID: 40001066 PMCID: PMC11863714 DOI: 10.1186/s12916-024-03786-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 11/19/2024] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Antibiotic therapy (ABT)-induced dysbiosis may affect the efficacy of immune checkpoint inhibitors (ICI) therapy. We investigated the association between ABT and real-world overall survival (rwOS) and progression-free survival (rwPFS) in patients with metastatic urothelial carcinoma (mUC) receiving ICI or cisplatin-based chemotherapy (CIS). METHODS Three thousand, one hundred seventy-nine patients were included from a nationwide electronic health record-derived de-identified database. Three-month landmark Kaplan-Meier methods and log-rank tests were used to estimate rwOS/PFS between treatment modalities based on ABT groups (stratified by exposure, timing, excretion mode, and administration route). Cox proportional models with time-varying coefficients were used to investigate the associations between ABT, treatment modality, and rwOS/PFS. RESULTS A total of 402 (27.1%) ICI and 655 (38.6%) CIS patients received ABT (p < 0.001). ICI receipt (OR 0.65, p < 0.001) and advanced age (OR 0.98, p < 0.001) were associated with lower ABT use. ICI exclusive findings included a negative correlation with rwOS in patients who received post-treatment initiated (ICI median: pre-13.2 vs post-7.9 vs none-13.3 months; p = 0.009), oral (median oral-9.6 vs none-13.3 months, p = 0.03), and renally cleared (median renal-9.9 vs none-13.3 months, p = 0.04) ABT. ABT's effect was negatively associated with rwOS in ICI patients within first 6 months (HR 1.36, 95% CI 1.107-1.74, p = 0.01) but not thereafter (p = 0.7). CONCLUSIONS This study identified a potential ICI-specific negative correlation between ABT and rwOS in patients with mUC, specifically those exposed to ABT pills and receipt before treatment initiation. These results emphasize the importance of antibiotic stewardship and continued investigation of the role of gut microbiome in mUC treatment efficacy.
Collapse
Affiliation(s)
- Avery Braun
- Department of Urologic Surgery, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, CA, 95817, USA.
| | - Mengying Deng
- Biostatistics and Bioinformatics, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Jill S Hasler
- Biostatistics and Bioinformatics, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Laura Bukavina
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
- Department of Urology, University Hospitals Cleveland Medical Center, Case Western Reserve School of Medicine, Cleveland, OH, 44106, USA
| | - Elizabeth Handorf
- Biostatistics and Bioinformatics, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Philip H Abbosh
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
- Department of Urology, Einstein Healthcare Network, Philadelphia, PA, 19141, USA
| |
Collapse
|
20
|
Fiala O, Buti S, Fujita K, de Liaño AG, Fukuokaya W, Kimura T, Yanagisawa T, Giannatempo P, Angel M, Mennitto A, Molina-Cerrillo J, Bourlon MT, Soares A, Takeshita H, Calabrò F, Ortega C, Kucharz J, Milella M, Seront E, Park SH, Tural D, Benedetti G, Ürün Y, Battelli N, Melichar B, Poprach A, Buchler T, Kopecký J, Conteduca V, Monteiro FSM, Massari F, Gupta S, Santoni M. Concomitant medications in patients with metastatic urothelial carcinoma receiving enfortumab vedotin: real-world data from the ARON-2 EV study. Clin Exp Metastasis 2025; 42:18. [PMID: 39976819 PMCID: PMC11842414 DOI: 10.1007/s10585-025-10335-4] [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: 12/02/2024] [Accepted: 02/10/2025] [Indexed: 02/23/2025]
Abstract
Patients with metastatic urothelial carcinoma (mUC) are typically elderly and often have other comorbidities that require the use of concomitant medications. In our study we evaluated the association of concomitant use of antibiotics (ATBs), proton pump inhibitors (PPIs), corticosteroids, statins, metformin and insulin with patient outcomes and we validated the prognostic role of a concomitant drug score in mUC patients treated with enfortumab vedotin (EV) monotherapy. Data from 436 patients enrolled in the ARON-2EV retrospective study were analyzed according to the concomitant medications used at baseline. Finally, the patients were stratified into three risk groups according to the concomitant drug score based on ATBs, corticosteroids and PPIs. Statistical analysis involved Fisher exact test, Kaplan-Meier method, log-rank test, and univariate/multivariate Cox proportional hazard regression models. Inferior survival outcomes were observed in ATB users compared to non-users (OS: 7.3 months, 95%CI 5.0 - 12.3 vs 13.7 months, 95%CI 12.2 - 47.3, p = 0.001; PFS: 5.1 months 95%CI 3.3 - 17.7 vs 8.3 months, 95%CI 7.1 - 47.3, p = 0.001) and also in corticosteroid users compared to non-users (OS: 8.4 months, 95%CI 6.6 - 10.0 vs 14.2 months, 95%CI 12.7 - 47.3, p < 0.001; PFS: 6.0 months 95%CI 4.6 - 7.9 vs 8.9 months, 95%CI 7.2 - 47.3, p = 0.004). In the Cox multivariate analysis, the concomitant drug score was a significant factor predicting both OS (HR = 1.32 [95% CI 1.03 - 1.68], p = 0.026) and PFS (HR = 1.23 [95% CI 1.01 - 1.51], p = 0.044). Our findings suggest detrimental impact of concomitant use of ATBs and corticosteroids on survival outcomes and the prognostic utility of the concomitant drug score in previously treated mUC patients receiving EV.
Collapse
Affiliation(s)
- Ondřej Fiala
- Department of Oncology and Radiotherapeutics, Faculty of Medicine and University Hospital in Pilsen, Charles University Prague, Alej Svobody 80, 304 60, Pilsen, Czech Republic.
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.
| | - Sebastiano Buti
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Kazutoshi Fujita
- Department of Urology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Alfonso Gómez de Liaño
- Department of Medical Oncology, Complejo Hospitalario Universitario Insular-Materno Infantil, Las Palmas, Spain
| | - Wataru Fukuokaya
- Department of Urology, The Jikei University School of Medicine, 3-19-18 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8471, Japan
| | - Takahiro Kimura
- Department of Urology, The Jikei University School of Medicine, 3-19-18 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8471, Japan
| | - Takafumi Yanagisawa
- Department of Urology, The Jikei University School of Medicine, 3-19-18 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8471, Japan
| | - Patrizia Giannatempo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| | - Martin Angel
- Clinical Oncology, Genitourinary Oncology Unit, Alexander Fleming Institute, Buenos Aires, Argentina
| | - Alessia Mennitto
- Department of Medical Oncology, Azienda Ospedaliera Universitaria "Maggiore Della Carit", Novara, Italy
| | | | - Maria T Bourlon
- Department of Hemato-Oncology, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Escuela de Medicina, Mexico-Universidad Panamericana, Mexico City, Mexico
| | - Andrey Soares
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Latin American Cooperative Oncology Group-LACOG, Porto Alegre, Brazil
| | - Hideki Takeshita
- Department of Urology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Fabio Calabrò
- Medical Oncology 1-IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Cinzia Ortega
- Dipartimento di Oncologia, Ospedale Michele E Pietro Ferrero-Verduno (CN) ASLCN2 Alba E, Bra, Italy
| | - Jakub Kucharz
- Department of Uro-Oncology, Maria Sklodowska-Curie National Research Institute of Oncology Warsaw, Warsaw, Poland
| | - Michele Milella
- Section of Innovation Biomedicine-Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University and Hospital Trust (AOUI) of Verona, 37134, Verona, Italy
| | - Emmanuel Seront
- Department of Medical Oncology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Se Hoon Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Deniz Tural
- Department of Medical Oncology, Koc University Medical Faculty, Istanbul, Türkiye
| | | | - Yüksel Ürün
- Department of Medical Oncology, Ankara University Faculty of Medicine, 06620, Ankara, Türkiye
| | | | - Bohuslav Melichar
- Department of Oncology, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic
| | - Alexandr Poprach
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, 656 53, Brno, Czech Republic
- Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
| | - Tomas Buchler
- Department of Oncology, Second Faculty of Medicine, Charles University and Motol University Hospital, V Úvalu 84, 150 06, Prague, Czech Republic
| | - Jindřich Kopecký
- Department of Oncology, University Hospital in Hradec Králové, Sokolská 581, 50005, Hradec Králové, Czech Republic
| | - Vincenza Conteduca
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy
| | | | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Shilpa Gupta
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Matteo Santoni
- Medical Oncology Unit, Macerata Hospital, Macerata, Italy
| |
Collapse
|
21
|
Wang N, Wu S, Huang L, Hu Y, He X, He J, Hu B, Xu Y, Rong Y, Yuan C, Zeng X, Wang F. Intratumoral microbiome: implications for immune modulation and innovative therapeutic strategies in cancer. J Biomed Sci 2025; 32:23. [PMID: 39966840 PMCID: PMC11837407 DOI: 10.1186/s12929-025-01117-x] [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: 09/09/2024] [Accepted: 01/09/2025] [Indexed: 02/20/2025] Open
Abstract
Recent advancements have revealed the presence of a microbiome within tumor tissues, underscoring the crucial role of the tumor microbiome in the tumor ecosystem. This review delves into the characteristics of the intratumoral microbiome, underscoring its dual role in modulating immune responses and its potential to both suppress and promote tumor growth. We examine state-of-the-art techniques for detecting and analyzing intratumoral bacteria, with a particular focus on their interactions with the immune system and the resulting implications for cancer prognosis and treatment. By elucidating the intricate crosstalk between the intratumoral microbiome and the host immune system, we aim to uncover novel therapeutic strategies that enhance the efficacy of cancer treatments. Additionally, this review addresses the existing challenges and future prospects within this burgeoning field, advocating for the integration of microbiome research into comprehensive cancer therapy frameworks.
Collapse
Affiliation(s)
- Na Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Si Wu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Lanxiang Huang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yue Hu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xin He
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jourong He
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ben Hu
- Center for Tumor Precision Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yaqi Xu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yuan Rong
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Chunhui Yuan
- Department of Laboratory Medicine, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430016, China.
| | - Xiantao Zeng
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Fubing Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, 430071, China.
| |
Collapse
|
22
|
Mafe AN, Büsselberg D. Microbiome Integrity Enhances the Efficacy and Safety of Anticancer Drug. Biomedicines 2025; 13:422. [PMID: 40002835 PMCID: PMC11852609 DOI: 10.3390/biomedicines13020422] [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: 01/22/2025] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
The intricate relationship between anticancer drugs and the gut microbiome influences cancer treatment outcomes. This review paper focuses on the role of microbiome integrity in enhancing the efficacy and safety of anticancer drug therapy, emphasizing the pharmacokinetic interactions between anticancer drugs and the gut microbiota. It explores how disruptions to microbiome composition, or dysbiosis, can alter drug metabolism, immune responses, and treatment side effects. By examining the mechanisms of microbiome disruption caused by anticancer drugs, this paper highlights specific case studies of drugs like cyclophosphamide, 5-fluorouracil, and irinotecan, and their impact on microbial diversity and clinical outcomes. The review also discusses microbiome-targeted strategies, including prebiotics, probiotics, postbiotics, and fecal microbiota transplantation (FMT), as promising interventions to enhance cancer treatment. Furthermore, the potential of microbiome profiling in personalizing therapy and integrating these interventions into clinical practice is explored. Finally, this paper proposes future research directions, including developing novel biomarkers and a deeper comprehension of drug-microbiome interactions, to respond to current gaps in knowledge and improve patient outcomes in cancer care.
Collapse
Affiliation(s)
- Alice N. Mafe
- Department of Biological Sciences, Faculty of Sciences, Taraba State University, Main Campus, Jalingo 660101, Taraba State, Nigeria;
| | - Dietrich Büsselberg
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha Metropolitan Area, Doha P.O. Box 22104, Qatar
| |
Collapse
|
23
|
Vaziri Y. Dietary influence on cancer progression: Gut health and genomic profiles. Curr Probl Cancer 2025; 54:101159. [PMID: 39615199 DOI: 10.1016/j.currproblcancer.2024.101159] [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: 06/15/2024] [Revised: 09/10/2024] [Accepted: 11/15/2024] [Indexed: 01/20/2025]
Abstract
This scholarly review comprehensively examines the connection between dietary habits, gut health, cancer prognosis, and genomic profiles. It emphasizes the crucial role of gut microbiota in mediating genomic changes and oncogenic processes through metabolic derivatives.It advocatеs for pеrsonalizеd nutrition stratеgiеs based on individual microbiomе and gеnomic profilеs and proposеs that customized diеtary intеrvеntions could play a crucial rolе in cancеr prеvеntion thеrapy. Thе article highlights thе influеncе of spеcific nutriеnts and such as diеtary fibеr and polyphеnols found in cеrtain foods and dеmonstrating thеir potеntial to altеr gеnе еxprеssions associatеd with inflammation and tumorigеnеsis. Thе rеviеw citеs rеcеnt studiеs that support thе idеa that diеtary modifications can influеncе gеnе rеgulation and thеrеby potеntially altеring cancеr progrеssion. Nevertheless, it calls for morе rigorous rеsеarch including longitudinal and randomizеd studies, to substantiatе thе еvidеncе nеcеssary for developing diеtary guidеlinеs tailorеd for cancеr patiеnts. Thе rеviеw еmphasizеs thе nееd for a multidisciplinary approach and highlight thе importancе of collaboration across thе fiеlds of nutrition gеnomics microbiology and oncology to improve cancеr trеatmеnts and patiеnt quality of lifе. It posits thе rеviеw as a cornеrstonе for a divеrsе audiеncе within thе scientific and mеdical communitimphasizing thе nеcеssity for ongoing rеsеarch in nutritional gеnomics which it dеpicts as a fiеld full of opportunitiеs to transform cancеr carе.
Collapse
Affiliation(s)
- Yashar Vaziri
- Department of Nutrition and Dietetics, Sarab branch, Islamic Azad University, Sarab, Iran.
| |
Collapse
|
24
|
Mondal R, Ritu RB, Kitaoka K, Azahar NM, Moniruzzaman M, Ogata S, Kiyoshige E, Tohara H, Kobayashi Y, Kashihara N, Naito T, Nakashima N, Tamura K, Nishimura K, Viera AJ, Yano Y. Oral microbiome alpha diversity and all-cause, cardiovascular, and non-cardiovascular mortality in US adults: Evidence from the NHANES 2009-2019. Atherosclerosis 2025; 401:119074. [PMID: 39644613 DOI: 10.1016/j.atherosclerosis.2024.119074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/24/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND AND AIMS Knowledge about the association between oral microbiome diversity within individuals and cardiovascular disease (CVD) and non-CVD mortality is scarce. Besides, variation by sex and racial and ethnic groups, and the potential mediators of these associations remain unclear. We aimed to investigate the associations of oral microbiome alpha diversity with all-cause, CVD, and non-CVD mortality, and the interaction effects of sex and racial and ethnic groups and potential mediators in the associations. METHODS The National Health and Nutrition Examination Survey (NHANES) is a population-based observational study, conducted periodically in Mexican American, Other Hispanic, Non-Hispanic (NH) White, NH Black, and other racial/ethnic participants. We linked 2009-12 survey data of 8199 adults to the mortality data until 2019. By analyzing RNA gene sequences from oral rinse samples, microbiome alpha diversity within individuals was assessed using operational taxonomic unit (OTU) richness. Potential mediators included obesity, diabetes mellitus, dyslipidemia, hypertension, and periodontitis. Multivariable Cox proportional hazards regression and causal mediation analysis were used. RESULTS Baseline mean ± standard deviation (SD) age was 42.1 ± 15.1 years. Over a median follow-up of 9.1 years, 405 all-cause mortality occurred (CVD, 105; non-CVD, 300). Each 1-SD increment in OTU richness was inversely associated with all-cause mortality (hazard ratio [HR] 0.92, 95 % confidence interval [CI] 0.90-0.95), CVD mortality (HR, 0.92; 95 % CI, 0.90-0.95), and non-CVD mortality (HR, 0.92; 95 % CI, 0.90-0.95). With evidence of significant racial and ethnic groups-interaction (p <0.05), these associations were evident in Mexican American, NH White, and others racial/ethnic participants. None of the potential mediators significantly mediated the associations of OTU richness with all-cause, CVD, and non-CVD mortality. CONCLUSIONS Lower oral microbiome alpha diversity is associated with higher risk for all-cause, CVD, and non-CVD mortality, and the associations are varied by racial and ethnic groups.
Collapse
Affiliation(s)
- Rajib Mondal
- Department of Preventive Medicine, NCD Epidemiology Research Center, Shiga University of Medical Science, Shiga, Japan; Department of Preventive Medicine and Epidemiology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Rani Baroi Ritu
- Department of Preventive Medicine, NCD Epidemiology Research Center, Shiga University of Medical Science, Shiga, Japan
| | - Kaori Kitaoka
- Department of Advanced Epidemiology, NCD Epidemiology Research Center, Shiga University of Medical Science, Shiga, Japan
| | - Nazar Mohd Azahar
- NCD Epidemiology Research Center, Shiga University of Medical Science, Shiga, Japan; Faculty of Health Sciences, Universiti Teknologi MARA, Cawangan Pulau Pinang, Kampus Bertam, Pulau Pinang, Malaysia
| | - Mohammad Moniruzzaman
- NCD Epidemiology Research Center, Shiga University of Medical Science, Shiga, Japan; Socio-Spatial Determinants of Health (SSDH) Laboratory, Population and Community Health Sciences Branch, Division of Intramural Research, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Soshiro Ogata
- Department of Preventive Medicine and Epidemiology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Eri Kiyoshige
- Department of Preventive Medicine and Epidemiology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Haruka Tohara
- Department of Dysphagia Rehabilitation, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Yusuke Kobayashi
- YCU Co-Creation Innovation Center, Yokohama City University, Yokohama, Japan
| | | | - Toshio Naito
- Department of General Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Naoki Nakashima
- Medical Information Center, Kyushu University Hospital, Japan
| | - Kosuke Tamura
- Socio-Spatial Determinants of Health (SSDH) Laboratory, Population and Community Health Sciences Branch, Division of Intramural Research, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Kunihiro Nishimura
- Department of Preventive Medicine and Epidemiology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Anthony J Viera
- Department of Family Medicine and Community Health, Duke University, NC, USA
| | - Yuichiro Yano
- Department of General Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan; Department of Family Medicine and Community Health, Duke University, NC, USA.
| |
Collapse
|
25
|
Malvezzi H, Cestari BA, Mendes H, Hernandes C, Podgaec S. Peritoneal fluid microbiota profile of patients with deep endometriosis. Microb Pathog 2025; 199:107244. [PMID: 39708979 DOI: 10.1016/j.micpath.2024.107244] [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: 08/15/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Endometriosis is a chronic gynecological disease that affects 10 % of reproductive-aged women and characterized by the presence of endometrial tissue outside the uterus. The disease is linked to a pro-inflammatory environment in the peritoneal fluid of patients, with high levels of cytokines, growth factors, and reactive oxygen species. Changes in the peritoneal fluid, such as altered immune cells and cytokines, can be linked to the immune balance in endometriosis. Immunological changes may be related to the presence of microorganisms in the peritoneal fluid that can activate Toll-like receptor (TLR) signaling and trigger an inflammatory response. A high diversity of TLRs has been found in women with endometriosis, and the presence of specific microorganisms in the fluid is suggested to be responsible for the activation of inflammasomes and inflammatory cytokines involved in the development of endometriosis. The present study was conducted at a hospital in southeastern Brazil to test this hypothesis, using a case-control design. Peritoneal fluid from 50 patients was used in this study. The case group consisted of 27 patients with endometriosis and the control group consisted of 23 patients without endometriosis. The samples were stored in a microbiome transport solution, and DNA was extracted and sent for genetic sequencing to identify the microorganisms present. The obtained sequencing reads were processed using a bioinformatics pipeline involving demultiplexing with the Illumina proprietary software, primer detection and removal, error evaluation, quality filtering, error removal using the Deblur software, amplicon sequence variants grouping, and chimera detection using the VSEARCH software. The sheer abundance of the microbiome made it challenging to discern any notable differences between the two groups. Nevertheless, we highlighted the prevalence of three primary bacteria in the peritoneal fluid from patients with endometriosis: Flavobacterium, Pseudomonas, and Bacillus. The results were established after a rigorous experimental design to eliminate potential contamination from extraction kits and handling. Our findings provide valuable insight into the pathogenesis of this disease and can be useful to understand how microbiota and immune system works in endometriosis.
Collapse
Affiliation(s)
- Helena Malvezzi
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi, São Paulo, 05651-901, Brazil.
| | - Bruna Azevedo Cestari
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi, São Paulo, 05651-901, Brazil
| | - Helen Mendes
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi, São Paulo, 05651-901, Brazil
| | - Camila Hernandes
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi, São Paulo, 05651-901, Brazil
| | - Sergio Podgaec
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi, São Paulo, 05651-901, Brazil; Departamento de Obstetricia e Ginecologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
26
|
Sanabani SS. Impact of Gut Microbiota on Lymphoma: New Frontiers in Cancer Research. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:e82-e89. [PMID: 39299827 DOI: 10.1016/j.clml.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/22/2024]
Abstract
The gut microbiome (GMB), which is made up of various microorganisms, plays a crucial role in maintaining the health of the host. Disruptions in this delicate ecosystem, known as microbial dysbiosis, have been linked to various diseases, including hematologic malignancies such as lymphoma. This review article explores the complex relationship between the GMB and the development of lymphoma and highlights its implications for diagnostic and therapeutic approaches. It discusses how GMB influences lymphoma development directly through the presence of certain microorganisms and indirectly through changes in the immune system. The clinical relevance of GMB is highlighted and its potential utility for diagnosis, predicting treatment outcomes and developing personalized therapeutic strategies for lymphoma patients is demonstrated. The review also looks at microbiome-targeted interventions such as fecal microbiome transplantation and dietary modification, which have shown promise for treating microbial dysbiosis and improving patient outcomes. In addition, it highlights the analytical challenges and the need for further research to fully elucidate the mechanistic functions of the GMB in the context of lymphoma. This review emphasizes the critical role of GMB in lymphomagenesis and its potential for the development of diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Sabri Saeed Sanabani
- Laboratory of Medical Investigation LIM 03, Hospital das Clínicas (HCFMU), School of Medicine, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
27
|
Salvestrini V, Conti G, D'Amico F, Cristiano G, Candela M, Cavo M, Turroni S, Curti A. Gut Microbiome as a Potential Marker of Hematologic Recovery Following Induction Therapy in Acute Myeloid Leukemia Patients. Cancer Med 2025; 14:e70501. [PMID: 39865898 PMCID: PMC11770270 DOI: 10.1002/cam4.70501] [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: 08/09/2024] [Revised: 11/27/2024] [Accepted: 12/01/2024] [Indexed: 01/28/2025] Open
Abstract
BACKGROUND The management of acute myeloid leukemia (AML) is hindered by treatment-related toxicities and complications, particularly cytopenia, which remains a leading cause of mortality. Given the pivotal role of the gut microbiota (GM) in hemopoiesis and immune regulation, we investigated its impact on hematologic recovery during AML induction therapy. METHODS We profiled the GM of 27 newly diagnosed adult AML patients using 16S rRNA amplicon sequencing and correlated it with key clinical parameters before and after induction therapy. RESULTS Our investigation revealed intriguing associations between the GM composition and crucial recovery indicators, including platelet, lymphocyte, and neutrophil counts, and identified early GM signatures predictive of improved hematologic recovery. Remarkably, patients demonstrating superior recovery had higher alpha diversity and enrichment in health-associated taxa belonging to the genera Faecalibacterium, Ruminococcus, Blautia, and Butyricimonas at diagnosis. CONCLUSIONS Despite certain study limitations, our findings suggest that evaluating GM features could serve as a potential marker for hematologic recovery. This preliminary work opens avenues for personalized risk assessment and interventions, possibly involving GM modulation tools, to optimize recovery in AML patients undergoing induction therapy and potentially enhancing overall outcomes in individuals with hematologic diseases.
Collapse
Affiliation(s)
- Valentina Salvestrini
- Istituto di Ematologia "Seràgnoli"IRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Gabriele Conti
- Human Microbiomics Unit, Department of Medical and Surgical SciencesUniversity of BolognaBolognaItaly
| | - Federica D'Amico
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and BiotechnologyUniversity of BolognaBolognaItaly
| | - Gianluca Cristiano
- Oncology and Haematology Research Area, Department of Medical and Surgical SciencesUniversity of BolognaBolognaItaly
| | - Marco Candela
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and BiotechnologyUniversity of BolognaBolognaItaly
| | - Michele Cavo
- Istituto di Ematologia "Seràgnoli"IRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
- Oncology and Haematology Research Area, Department of Medical and Surgical SciencesUniversity of BolognaBolognaItaly
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and BiotechnologyUniversity of BolognaBolognaItaly
| | - Antonio Curti
- Istituto di Ematologia "Seràgnoli"IRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| |
Collapse
|
28
|
Fang X, Wang Z, Chen Q, Du Y, Sun H, Liu H, Feng Y, Li Z, Teng T, Shi B. Protective effect of the branched short-chain fatty acid isobutyrate on intestinal damage in weaned piglets through intestinal microbiota remodeling. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:1556-1568. [PMID: 39412364 DOI: 10.1002/jsfa.13930] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 01/14/2025]
Abstract
BACKGROUND Postweaning intestinal damage in piglets is a challenging issue in the livestock industry. Short-chain fatty acids (SCFAs) are important metabolic products of the gut microbiota and are widely recognized for their role in maintaining normal colonic function and regulating the intestinal immune system. However, the effects of branched short-chain fatty acid (BSCFA) isobutyrate on intestinal health remain largely unknown. This study aims to explore the potential of isobutyrate for alleviating postweaning intestinal damage. RESULTS This study indicates that isobutyrate can alleviate diarrhea in weaned piglets, enhance their growth performance, and optimize the gut microbiota. This is mainly achieved through increasing the relative abundance of probiotic bacteria such as Lactobacillus, Megasphaera, and Prevotellaceae_UCG-003, while concurrently reducing the relative abundance of potentially harmful bacteria such as Clostridium_sensu_stricto-1 and Escherichia-Shigella. It promotes the production of SCFAs, including acetate, isobutyrate, and butyrate. Furthermore, it activates G-protein-coupled receptors (GPR43/109A), inhibits the TLR4/MyD88 signaling pathway, strengthens the intestinal barrier function, and regulates the expression of related cytokines. CONCLUSION In summary, exogenous isobutyrate can be considered a promising feed additive for improving the intestinal microbiota and regulating intestinal health in piglets. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xiuyu Fang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Zhengyi Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Qinrui Chen
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Yongqing Du
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Haowen Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Haiyang Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Ye Feng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Zhongyu Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Teng Teng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Baoming Shi
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| |
Collapse
|
29
|
Emadi R, Saki S, Yousefi P, Tabibzadeh A. A Perspective on Lung Cancer and Lung Microbiome: Insight on Immunity. Immun Inflamm Dis 2025; 13:e70145. [PMID: 39887959 PMCID: PMC11783403 DOI: 10.1002/iid3.70145] [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: 05/02/2024] [Revised: 01/03/2025] [Accepted: 01/19/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Although the carcinogenic potential of microbes has long been recognized, their significance may have been underestimated. Currently, the connection between microbiota and cancer is under extensive research. The lung microbiota may serve as a proxy for the state of lung health based on its crucial role in preserving lung hemostasis. OBJECTIVES This review tried to outline the state of our understanding of the contribution of lung microbiome and lung cancer. METHODS A literature search was performed using PubMed, Google Scholar, and Scopus databases for recent research focusing on the development and possible pathogenesis of lung microbiome and lung cancer. RESULTS Early research on lung cancer indicated that dysbiosis significantly impacted the development and spread of the tumor. As a result of these findings, the study of the lung microbiota as a possible therapeutic target and diagnostic marker has accelerated. Early-stage disease diagnostic biomarkers could be represented as microbiota profiles. Additionally, the microbiome is involved in anticancer therapy. There are limited studies on lung microbiota, and most microbiome studies commonly concentrate on the gut microbiota. A proper understanding of lung microbiota can have several potential therapeutic approaches. Therefore, more studies in this field may initiate remarkable advancements in microbiome-dependent treatment. CONCLUSION Convincing data from studies on both humans and animals indicates that the microbiota might play a role in cancer initiation, influenced by internal and environmental factors of the host. Notably, the lung harbors its microbiome, as do lung cancers. In general view, it seems microbiome diversity in lung cancer patients is reduced. Meanwhile, some genera were increased in lung cancer patients in comparison with a noncancerous population (such as Streptococcus genus), and some of them were decreased (Granulicatella adiacens, G. adiacens). Furthermore, research on the microbiome-carcinogenesis relationship is still in its infancy, and much remains to be fully understood.
Collapse
Affiliation(s)
- Reza Emadi
- Department of Medical Laboratory Sciences, Faculty of Medical SciencesIslamic Azad University, Arak BranchArakIran
| | - Sasan Saki
- Department of Medical Laboratory Sciences, Faculty of Medical SciencesIslamic Azad University, Arak BranchArakIran
| | - Parastoo Yousefi
- Department of Virology, School of MedicineIran University of Medical SciencesTehranIran
| | - Alireza Tabibzadeh
- Department of Medical Laboratory Sciences, Faculty of Medical SciencesIslamic Azad University, Arak BranchArakIran
- Applied Neuroscience Research CenterIslamic Azad University, Arak BranchArakIran
| |
Collapse
|
30
|
Huang W, Jiang T, He J, Ruan J, Wu B, Tao R, Xu P, Wang Y, Chen R, Wang H, Yang Q, Zhang K, Jin L, Sun D, You J. Modulation of Intestinal Flora: a Novel Immunotherapeutic Approach for Enhancing Thyroid Cancer Treatment. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10471-z. [PMID: 39890752 DOI: 10.1007/s12602-025-10471-z] [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] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Over the past 3 years, there has been a growing interest in clinical research regarding the potential involvement of intestinal flora in thyroid cancer (TC). This review delves into the intricate connection between intestinal flora and TC, focusing on the particular intestinal flora that is directly linked to the disease and identifying which may be able to predict potential microbial markers of TC. In order to shed light on the inflammatory pathways connected to the onset of TC, we investigated the impact of intestinal flora on immune modulation and the connection between chronic inflammation when investigating the role of intestinal flora in the pathogenesis of TC. Furthermore, the potential role of intestinal flora metabolites in the regulation of thyroid function was clarified by exploring the effects of short-chain fatty acids and lipopolysaccharide on thyroid hormone synthesis and metabolism. Based on these findings, we further explore the effects of probiotics, prebiotics, postbiotics, vitamins, and trace elements.
Collapse
Affiliation(s)
- Weiqiang Huang
- Department of General Surgery, The First People's Hospital of Jiashan, Jiashan Hospital Afliated of Jiaxing University, Jiaxing, 314100, China
| | - Tao Jiang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jiaxuan He
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jing Ruan
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Baihui Wu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Runchao Tao
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Peiye Xu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Yongpan Wang
- Department of General Surgery, The First People's Hospital of Jiashan, Jiashan Hospital Afliated of Jiaxing University, Jiaxing, 314100, China
| | - Rongbing Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, SAR 999077, China
| | - Hanbing Wang
- The University of Hong Kong School of Biomedical Sciences, Hong Kong, 999077, SAR, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Kun Zhang
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China.
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China.
| | - Jinfeng You
- Department of Obstetrics, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China.
| |
Collapse
|
31
|
Song JH, Lim KM, Yoo SH, Kim GD, Shin HS, Park S, Lim MY, Lee SY. Effects of Limosilactobacillus fermentum KBL375 on Immune Enhancement and Gut Microbiota Composition in Cyclophosphamide-Induced Immunosuppressed Mice. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10463-z. [PMID: 39885060 DOI: 10.1007/s12602-025-10463-z] [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] [Accepted: 01/17/2025] [Indexed: 02/01/2025]
Abstract
This study evaluated the immune-enhancing efficacy of Limosilactobacillus fermentum KBL375 isolated from the feces of healthy Koreans. KBL375-treated splenocytes showed enhancement of cytotoxicity against YAC-1 cells, the target of natural killer (NK) cells, with an increase in CD335, granzyme B, perforin, and interferon-gamma (IFN-γ). Oral administration of KBL375 in mice with cyclophosphamide (CP)-induced immunosuppression improved body weight and immune functions, including immune organ indices, lymphocyte proliferations, and immunoglobulin (Ig) A levels. Notably, KBL375 increased NK cell cytotoxicity and proportion in immunosuppressed mice. Perforin/IFN-γ expression levels, which indicated NK cell activation, were also increased in KBL375-treated mice. Furthermore, KBL375 led to an increase in beneficial microbes, such as Bifidobacterium, in the gut microbiome of immunosuppressed mice, fostering a favorable intestinal microbial environment. These comprehensive results suggest that KBL375 exhibits potent immune regulatory functions and positively influences the gut microbiota, implying its potential as a probiotic agent for immune enhancement.
Collapse
Affiliation(s)
- Ju Hye Song
- Division of Food Functionality Research, Korea Food Research Institute, 245, Nongsaengmyeong-Ro, Iseo-Myeon, Wanju-Gun, 55365, Jeollabuk-Do, Republic of Korea
- Department of Food Biotechnology, Korea, University of Science and Technology, Daejeon, Republic of Korea
| | - Kyung Min Lim
- Division of Food Functionality Research, Korea Food Research Institute, 245, Nongsaengmyeong-Ro, Iseo-Myeon, Wanju-Gun, 55365, Jeollabuk-Do, Republic of Korea
- Department of Food Biotechnology, Korea, University of Science and Technology, Daejeon, Republic of Korea
| | - Sang Hyuk Yoo
- Division of Food Functionality Research, Korea Food Research Institute, 245, Nongsaengmyeong-Ro, Iseo-Myeon, Wanju-Gun, 55365, Jeollabuk-Do, Republic of Korea
- Department of Food Biotechnology, Korea, University of Science and Technology, Daejeon, Republic of Korea
| | - Gun-Dong Kim
- Division of Food Functionality Research, Korea Food Research Institute, 245, Nongsaengmyeong-Ro, Iseo-Myeon, Wanju-Gun, 55365, Jeollabuk-Do, Republic of Korea
| | - Hee Soon Shin
- Division of Food Functionality Research, Korea Food Research Institute, 245, Nongsaengmyeong-Ro, Iseo-Myeon, Wanju-Gun, 55365, Jeollabuk-Do, Republic of Korea
- Department of Food Biotechnology, Korea, University of Science and Technology, Daejeon, Republic of Korea
| | | | - Mi Young Lim
- Division of Food Functionality Research, Korea Food Research Institute, 245, Nongsaengmyeong-Ro, Iseo-Myeon, Wanju-Gun, 55365, Jeollabuk-Do, Republic of Korea.
| | - So-Young Lee
- Division of Food Functionality Research, Korea Food Research Institute, 245, Nongsaengmyeong-Ro, Iseo-Myeon, Wanju-Gun, 55365, Jeollabuk-Do, Republic of Korea.
- Department of Food Biotechnology, Korea, University of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
32
|
Sapkota H, Dasgupta S, Roy B, Pathan EK. Human Commensal Bacteria: Next-generation Pro- and Post-biotics for Anticancer Therapy. Front Biosci (Elite Ed) 2025; 17:26809. [PMID: 40150985 DOI: 10.31083/fbe26809] [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: 10/01/2024] [Revised: 12/08/2024] [Accepted: 12/23/2024] [Indexed: 03/29/2025]
Abstract
Cancer is a common, deadly disease with an unknown etiology. Meanwhile, current therapeutic options possess significant risks. However, probiotic bacteria and their metabolites have been reported to have antiproliferative and apoptotic effects on cancer cells. Therefore, because of their selective specificity and lack of treatment-associated comorbidities, these bacteria and their metabolites could be potential alternatives to conventional chemical and radiation therapies. Given their superior immunomodulatory and anti-cancer effects and lack of side effects, commensal bacteria derived from healthy humans are currently used as next-generation probiotics. This review summarizes current findings on these probiotic properties and anti-cancer activities of healthy human commensal bacteria. Additionally, the review focuses on small metabolites, proteins, and enzymes secreted by human commensal bacteria for their therapeutic applications against cancer. Further, utilizing a protein engineering strategy to reduce the toxicity of L-asparaginase, an enzyme-based anti-leukemia drug used for the last forty years, is also discussed. A possible workflow outline for isolating, identifying, screening, and characterizing human commensal bacterial strains for their therapeutic applications in cancer treatment is also proposed. This review emphasizes the need to explore various human commensal bacteria, not just mainstream lactic acid bacteria, for novel cancer therapeutics that provide multiple health benefits.
Collapse
Affiliation(s)
- Himal Sapkota
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), 412115 Pune, Maharashtra, India
| | - Subrata Dasgupta
- RIKEN Center for Biosystems Dynamics Research, 230-0045 Yokohama, Kanagawa, Japan
| | - Bishnudeo Roy
- Department of Biosciences and Technology, MIT World Peace University, 411038 Pune, Maharashtra, India
| | - Ejaj K Pathan
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), 412115 Pune, Maharashtra, India
| |
Collapse
|
33
|
Al-Matouq J, Al-Ghafli H, Alibrahim NN, Alsaffar N, Radwan Z, Ali MD. Unveiling the Interplay Between the Human Microbiome and Gastric Cancer: A Review of the Complex Relationships and Therapeutic Avenues. Cancers (Basel) 2025; 17:226. [PMID: 39858007 PMCID: PMC11763844 DOI: 10.3390/cancers17020226] [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: 11/25/2024] [Revised: 12/23/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
The human microbiota plays a crucial role in maintaining overall health and well-being. The gut microbiota has been implicated in developing and progressing various diseases, including cancer. This review highlights the related mechanisms and the compositions that influence cancer pathogenesis with a highlight on gastric cancer. We provide a comprehensive overview of the mechanisms by which the microbiome influences cancer development, progression, and response to treatment, with a focus on identifying potential biomarkers for early detection, prevention strategies, and novel therapeutic interventions that leverage microbiome modulation. This comprehensive review can guide future research and clinical practices in understanding and harnessing the microbiome to optimize gastric cancer therapies.
Collapse
Affiliation(s)
- Jenan Al-Matouq
- Department of Medical Laboratory Sciences, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam 34222, Saudi Arabia; (H.A.-G.); (N.N.A.); (N.A.); (Z.R.)
| | - Hawra Al-Ghafli
- Department of Medical Laboratory Sciences, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam 34222, Saudi Arabia; (H.A.-G.); (N.N.A.); (N.A.); (Z.R.)
| | - Noura N. Alibrahim
- Department of Medical Laboratory Sciences, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam 34222, Saudi Arabia; (H.A.-G.); (N.N.A.); (N.A.); (Z.R.)
| | - Nida Alsaffar
- Department of Medical Laboratory Sciences, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam 34222, Saudi Arabia; (H.A.-G.); (N.N.A.); (N.A.); (Z.R.)
| | - Zaheda Radwan
- Department of Medical Laboratory Sciences, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam 34222, Saudi Arabia; (H.A.-G.); (N.N.A.); (N.A.); (Z.R.)
| | - Mohammad Daud Ali
- Department of Pharmacy, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam 34222, Saudi Arabia;
| |
Collapse
|
34
|
Monti E, Vianello C, Leoni I, Galvani G, Lippolis A, D’Amico F, Roggiani S, Stefanelli C, Turroni S, Fornari F. Gut Microbiome Modulation in Hepatocellular Carcinoma: Preventive Role in NAFLD/NASH Progression and Potential Applications in Immunotherapy-Based Strategies. Cells 2025; 14:84. [PMID: 39851512 PMCID: PMC11764391 DOI: 10.3390/cells14020084] [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: 12/23/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a heterogeneous tumor associated with several risk factors, with non-alcoholic fatty liver disease (NAFLD) emerging as an important cause of liver tumorigenesis. Due to the obesity epidemics, the occurrence of NAFLD has significantly increased with nearly 30% prevalence worldwide. HCC often arises in the background of chronic liver disease (CLD), such as nonalcoholic steatohepatitis (NASH) and cirrhosis. Gut microbiome (GM) alterations have been linked to NAFLD progression and HCC development, with several investigations reporting a crucial role for the gut-liver axis and microbial metabolites in promoting CLD. Moreover, the GM affects liver homeostasis, energy status, and the immune microenvironment, influencing the response to immunotherapy with interesting therapeutic implications. In this review, we summarize the main changes in the GM and derived metabolites (e.g., short-chain fatty acids and bile acids) occurring in HCC patients and influencing NAFLD progression, emphasizing their potential as early diagnostic biomarkers and prognostic tools. We discuss the weight loss effects of diet-based interventions and healthy lifestyles for the treatment of NAFLD patients, highlighting their impact on the restoration of the intestinal barrier and GM structure. We also describe encouraging preclinical findings on the modulation of GM to improve liver functions in CLD, boost the antitumor immune response (e.g., probiotic supplementations or anti-hypercholesterolemic drug treatment), and ultimately delay NAFLD progression to HCC. The development of safe and effective strategies that target the gut-liver axis holds promise for liver cancer prevention and treatment, especially if personalized options will be considered.
Collapse
Affiliation(s)
- Elisa Monti
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Clara Vianello
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Ilaria Leoni
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Giuseppe Galvani
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Annalisa Lippolis
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
| | - Federica D’Amico
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.D.); (S.R.); (S.T.)
| | - Sara Roggiani
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.D.); (S.R.); (S.T.)
- Human Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Claudio Stefanelli
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.D.); (S.R.); (S.T.)
- IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy
| | - Francesca Fornari
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
- IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
35
|
Zhou X, Wu Y, Zhu Z, Lu C, Zhang C, Zeng L, Xie F, Zhang L, Zhou F. Mucosal immune response in biology, disease prevention and treatment. Signal Transduct Target Ther 2025; 10:7. [PMID: 39774607 PMCID: PMC11707400 DOI: 10.1038/s41392-024-02043-4] [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: 06/01/2024] [Revised: 09/05/2024] [Accepted: 10/27/2024] [Indexed: 01/11/2025] Open
Abstract
The mucosal immune system, as the most extensive peripheral immune network, serves as the frontline defense against a myriad of microbial and dietary antigens. It is crucial in preventing pathogen invasion and establishing immune tolerance. A comprehensive understanding of mucosal immunity is essential for developing treatments that can effectively target diseases at their entry points, thereby minimizing the overall impact on the body. Despite its importance, our knowledge of mucosal immunity remains incomplete, necessitating further research. The outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has underscored the critical role of mucosal immunity in disease prevention and treatment. This systematic review focuses on the dynamic interactions between mucosa-associated lymphoid structures and related diseases. We delve into the basic structures and functions of these lymphoid tissues during disease processes and explore the intricate regulatory networks and mechanisms involved. Additionally, we summarize novel therapies and clinical research advances in the prevention of mucosal immunity-related diseases. The review also addresses the challenges in developing mucosal vaccines, which aim to induce specific immune responses while maintaining tolerance to non-pathogenic microbes. Innovative therapies, such as nanoparticle vaccines and inhalable antibodies, show promise in enhancing mucosal immunity and offer potential for improved disease prevention and treatment.
Collapse
Affiliation(s)
- Xiaoxue Zhou
- School of Medicine, Hangzhou City University, Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yuchen Wu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhipeng Zhu
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Chu Lu
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Chunwu Zhang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linghui Zeng
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Feng Xie
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
36
|
Guo L, An T. Investigating the correlation between gut microbiota and prostate cancer through a two-sample Mendelian randomization analysis. Medicine (Baltimore) 2025; 104:e41141. [PMID: 40184142 PMCID: PMC11709207 DOI: 10.1097/md.0000000000041141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/25/2024] [Accepted: 12/12/2024] [Indexed: 04/05/2025] Open
Abstract
Previous studies in observational epidemiology have suggested a potential correlation between the gastrointestinal tract microbiota and prostate cancer. However, the causal relationship between the 2 remains uncertain, our objective was to thoroughly examine the influence of the gut microbiome on the progression of prostate cancer. In this study, we focused on investigating the gut microbiome as an exposure factor, specifically analyzing data from the MiBioGen consortium, which had a substantial sample size of 18,340 participants. As our disease outcome, we utilized prostate cancer data from the FinnGen genome-wide association study, which involved 13,216 participants. To establish causal relationships, we conducted a comprehensive Mendelian randomization analysis employing multiple methods, including inverse variance-weighted, Mendelian randomization-Egger, maximum likelihood, and weighted median approaches. Additionally, we performed sensitivity analysis to address issues such as heterogeneity and horizontal pleiotropy, ensuring the robustness of our findings. The results obtained through inverse variance-weighted analysis revealed that certain microbial groups exhibited a protective effect on prostate cancer. Specifically, the phylum Verrucomicrobia, particularly the family Rikenellaceae, and the genera Anaerotruncus, Eisenbergiella, Olsenella, and Parabacteroides were found to have a beneficial impact. Conversely, the class Bacilli, class Erysipelotrichia, order Erysipelotrichales, order Lactobacillales, family Erysipelotrichaceae, and the genera Marvinbryantia, Romboutsia, Ruminococcaceae UCG002, and Sutterella had an adverse influence on prostate cancer. The sensitivity analysis did not reveal any such outliers, further strengthening the validity of our results. To summarize, a cause-and-effect connection was discovered between various types and prostate cancer. Nevertheless, additional randomized controlled experiments are required for validation.
Collapse
Affiliation(s)
- Lingyu Guo
- Department of Urology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Tian An
- Department of Dermatology and Plastic Surgery, The Second Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xianyang, China
| |
Collapse
|
37
|
Deng J, Sun C, Xu G, Wang B, Tzortzopoulou E, Deng D, Giovannetti E. The Oral Microbiome and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1472:151-170. [PMID: 40111691 DOI: 10.1007/978-3-031-79146-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
There is growing evidence suggesting a strong association between members of the oral microbiota and various types of cancer, including oral cancer, colorectal cancer, esophageal squamous cell carcinoma, and pancreatic cancer. Periodontal diseases closely associated with pathogenic bacteria in the oral cavity have been shown to be correlated with the occurrence and development of cancers. Among the periodontal disease-associated bacteria in the oral cavity, two prominent oral pathogens, Porphyromonas gingivalis and Fusobacterium nucleatum, have been found to promote tumor cell proliferation, invasion, and migration, as well as to inhibit immune cell function, thereby facilitating tumor progression. The presence of other oral pathogenic bacteria, such as Treponema denticola, Tannerella forsythia, Parvimonas micra, and Aggregatibacter actinomycetemcomitans, has also been found to be associated with cancer worsening. Oral commensal bacteria play a crucial role in maintaining the normal oral homeostasis. However, the relationship between oral commensal bacteria and the occurrence and development of cancers remains controversial. Some studies suggest an increase in oral commensal bacteria during tumor development, while others suggest an association of certain commensal bacteria with lower tumor risk. The microbiota can significantly alter responses and toxicity to various forms of cancer treatment through interactions with the human body, thereby influencing disease progression. In this chapter, we provide a concise overview of current understanding of the role of the oral microbiota in cancer.
Collapse
Affiliation(s)
- Juan Deng
- Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Chen Sun
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Geng Xu
- Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Bing Wang
- Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| | - Eleni Tzortzopoulou
- Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Fondazione Pisana per la Scienza, Pisa, Italy
| |
Collapse
|
38
|
Yu Z, Wang Q, Wang Z, Liu S, Xia T, Duan C, Liu Y, Ding X, Chen S, Yu T, You R, Chen M, Huang P. Lachnoclostridium intestinal flora is associated with immunotherapy efficacy in nasopharyngeal carcinoma. Head Neck 2025; 47:269-281. [PMID: 39135356 DOI: 10.1002/hed.27917] [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: 05/29/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Effective biomarkers for assessing anti-PD-1/PD-L1 therapy efficacy in patients with nasopharyngeal carcinoma (NPC) are still lacking. The human gut microbiota has been shown to influence clinical response to anti-PD-1/PD-L1 therapy in many cancers. However, the relationship between the gut microbiota and the efficacy of immunotherapy in patients with nasopharyngeal carcinoma has not been determined. METHODS We conducted a prospective study in which fecal and blood samples from patients with NPC were subjected to 16S rDNA sequencing and survival analysis. To investigate potential differences in the gut microbiome between these groups and to identify potential biomarkers indicative of immunotherapy efficacy, patients were categorized into two groups according to their clinical response to immunotherapy, the responder group (R group) and the non-responder group (NR group). Progression-free survival (PFS) between these subgroups was analyzed using Kaplan-Meier survival analysis with the log-rank test. Additionally, we performed univariate and multivariate analyses to evaluate prognostic factors. Finally, we carried out non-targeted metabolomics to examine the metabolic effects associated with the identified microbiome. RESULTS Our 16S rDNA sequencing results showed that the abundance of Lachnoclostridium was higher in the NR group than in the R group (p = 0.003), and alpha diversity analysis showed that the abundance of microbiota in the NR group was higher than that in the R group (p = 0.050). Patients with a lower abundance of Lachnoclostridium had better PFS (p = 0.048). Univariate (p = 0.017) and multivariate analysis (p = 0.040) showed that Lachnoclostridium was a predictor of PFS. Non-targeted metabolomics analysis revealed that Lachnoclostridium affects the efficacy of immunotherapy through the usnic acid. CONCLUSIONS High abundance of Lachnoclostridium predicts poor prognosis in patients with NPC receiving immunotherapy.
Collapse
Affiliation(s)
- Zikun Yu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qin Wang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zimeng Wang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Sihan Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tianliang Xia
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chongyang Duan
- Department of Biostatistics, School of Public Health, Southern Medical University, Guangzhou, China
| | - Youping Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Nasopharyngeal Cancer Prevention Center, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Xi Ding
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Siyuan Chen
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tao Yu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rui You
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Nasopharyngeal Cancer Prevention Center, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Mingyuan Chen
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Nasopharyngeal Cancer Prevention Center, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Peiyu Huang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
39
|
Palkovsky M, Modrackova N, Neuzil-Bunesova V, Liberko M, Soumarova R. The Bidirectional Impact of Cancer Radiotherapy and Human Microbiome: Microbiome as Potential Anti-tumor Treatment Efficacy and Toxicity Modulator. In Vivo 2025; 39:37-54. [PMID: 39740900 PMCID: PMC11705129 DOI: 10.21873/invivo.13803] [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: 07/29/2024] [Revised: 10/02/2024] [Accepted: 10/14/2024] [Indexed: 01/02/2025]
Abstract
Microbiome and radiotherapy represent bidirectionally interacting entities. The human microbiome has emerged as a pivotal modulator of the efficacy and toxicity of radiotherapy; however, a reciprocal effect of radiotherapy on microbiome composition alterations has also been observed. This review explores the relationship between the microbiome and extracranial solid tumors, particularly focusing on the bidirectional impact of radiotherapy on organ-specific microbiome. This article aims to provide a systematic review on the radiotherapy-induced microbial alteration in-field as well as in distant microbiomes. In this review, particular focus is directed to the oral and gut microbiome, its role in the development and progression of cancer, and how it is altered throughout radiotherapy. This review concludes with recommendations for future research, such as exploring microbiome modification to optimize radiotherapy-induced toxicities or enhance its anti-cancer effects.
Collapse
Affiliation(s)
- Martin Palkovsky
- Department of Oncology, University Hospital Kralovske Vinohrady, Prague, Czech Republic;
- Charles University, Third Faculty of Medicine, Department of Oncology, Prague, Czech Republic
| | - Nikol Modrackova
- Czech University of Life Sciences Prague, Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Prague, Czech Republic
| | - Vera Neuzil-Bunesova
- Czech University of Life Sciences Prague, Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Prague, Czech Republic
| | - Marian Liberko
- Department of Oncology, University Hospital Kralovske Vinohrady, Prague, Czech Republic
- Charles University, Third Faculty of Medicine, Department of Oncology, Prague, Czech Republic
| | - Renata Soumarova
- Department of Oncology, University Hospital Kralovske Vinohrady, Prague, Czech Republic
- Charles University, Third Faculty of Medicine, Department of Oncology, Prague, Czech Republic
| |
Collapse
|
40
|
Huang Y, Chen Z, Shen G, Fang S, Zheng J, Chi Z, Zhang Y, Zou Y, Gan Q, Liao C, Yao Y, Kong J, Fan X. Immune regulation and the tumor microenvironment in anti-PD-1/PDL-1 and anti-CTLA-4 therapies for cancer immune evasion: A bibliometric analysis. Hum Vaccin Immunother 2024; 20:2318815. [PMID: 38419524 PMCID: PMC11789735 DOI: 10.1080/21645515.2024.2318815] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/26/2024] [Accepted: 02/11/2024] [Indexed: 03/02/2024] Open
Abstract
This study aims to conduct a bibliometric analysis, employing visualization tools to examine literature pertaining to tumor immune evasion related to anti-CTLA-4 and anti-PD-1/PD-L1 therapy from 1999 to 2022. A special emphasis is placed on the interplay between tumor microenvironment, signaling pathways, immune cells and immune evasion, with data sourced from the Web of Science core collection (WoSCC). Advanced tools, including VOSviewer, Citespace, and Scimago Graphica, were utilized to analyze various parameters, such as co-authorship/co-citation patterns, regional contributions, journal preferences, keyword co-occurrences, and significant citation bursts. Out of 4778 publications reviewed, there was a marked increase in research focusing on immune evasion, with bladder cancer being notably prominent. Geographically, China, the USA, and Japan were the leading contributors. Prestigious institutions like MD Anderson Cancer Center, Harvard Medical School, Fudan University, and Sun Yat Sen University emerged as major players. Renowned journals in this domain included Frontiers in Immunology, Cancers, and Frontiers in Oncology. Ehen LP and Wang W were identified as prolific authors on this topic, while Topalian SL stood out as one of the most cited. Research current situation is notably pivoting toward challenges like immunotherapy resistance and the intricate signaling pathways driving drug resistance. This bibliometric study seeks to provide a comprehensive overview of past and current research trends, emphasizing the potential role of tumor microenvironment, signaling pathways and immune cells in the context of immune checkpoint inhibitors (ICIs) and tumor immune evasion.
Collapse
Affiliation(s)
- Yi Huang
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Zhijian Chen
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Gang Shen
- Department of Urology, DUSHU Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Shuogui Fang
- Department of Radiotherapy, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
| | - Junjiong Zheng
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Zepai Chi
- Department of urology, Shantou Central Hospital, Shantou, China
| | - Yuanfeng Zhang
- Department of urology, Shantou Central Hospital, Shantou, China
| | - Yitong Zou
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Qinghua Gan
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Chengxiao Liao
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Yuhui Yao
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Jianqiu Kong
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Xinxiang Fan
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen University, Guangzhou, P. R. China
| |
Collapse
|
41
|
Lefler DS, Manobianco SA, Bashir B. Immunotherapy resistance in solid tumors: mechanisms and potential solutions. Cancer Biol Ther 2024; 25:2315655. [PMID: 38389121 PMCID: PMC10896138 DOI: 10.1080/15384047.2024.2315655] [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: 07/24/2023] [Accepted: 02/04/2024] [Indexed: 02/24/2024] Open
Abstract
While the emergence of immunotherapies has fundamentally altered the management of solid tumors, cancers exploit many complex biological mechanisms that result in resistance to these agents. These encompass a broad range of cellular activities - from modification of traditional paradigms of immunity via antigen presentation and immunoregulation to metabolic modifications and manipulation of the tumor microenvironment. Intervening on these intricate processes may provide clinical benefit in patients with solid tumors by overcoming resistance to immunotherapies, which is why it has become an area of tremendous research interest with practice-changing implications. This review details the major ways cancers avoid both natural immunity and immunotherapies through primary (innate) and secondary (acquired) mechanisms of resistance, and it considers available and emerging therapeutic approaches to overcoming immunotherapy resistance.
Collapse
Affiliation(s)
- Daniel S. Lefler
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven A. Manobianco
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Babar Bashir
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
42
|
Yang Y, Shen J, Deng P, Chen P. Mechanism investigation of Forsythoside A against esophageal squamous cell carcinoma in vitro and in vivo. Cancer Biol Ther 2024; 25:2380023. [PMID: 39046082 PMCID: PMC11271126 DOI: 10.1080/15384047.2024.2380023] [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: 12/29/2023] [Revised: 06/21/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024] Open
Abstract
CONTEXT Forsythoside A (FSA) was extracted from Forsythia suspensa, a traditional Chinese medicine, which has been demonstrated to exert anti-inflammatory, antibacterial, and other pharmacological effects. However, the anticancer effect of FSA in esophageal squamous cell carcinoma (ESCC) has not been documented. OBJECTIVE The present study aimed to elucidate the mechanism of FSA against ESCC. MATERIALS AND METHODS Network pharmacology and molecular docking were employed to predict the mechanism. FSA was utilized to treat ESCC cell lines KYSE450 and KYSE30, followed by CCK-8 assay, cell cloning formation assay, flow cytometry, Western blot, RNA-seq analysis, and subsequent in vivo experiments. RESULTS Network pharmacology and molecular docking predicted that the therapeutic effect of FSA in ESCC is mediated through proteins such as BCL2 and BAX, influencing KEGG pathways associated with apoptosis. In vitro experiments showed that FSA inhibited cell proliferation and plate clone formation, promoted cell apoptosis and impacted the cell cycle distribution of G2/M phase by regulating BCL2, BAX, and p21. Further RNA-seq in KYSE450 cells showed that FSA regulated the expression of 223 genes, specifically affecting the biological process of epidermal development. In vivo experiments showed that gastric administration of FSA resulted in notable reductions in both tumor volume and weight by regulating BCL2, BAX, and p21. 16S rRNA sequencing showed that FSA led to significant changes of beta diversity. Abundance of 11 specific bacterial taxa were considerably changed following administration of FSA. CONCLUSIONS This study presents a novel candidate drug against ESCC and establishes a foundation for future clinical application.
Collapse
Affiliation(s)
- Yingying Yang
- School of Life Sciences, Zhengzhou Normal University, Zhengzhou, People’s Republic of China
| | - Junru Shen
- School of Life Sciences, Zhengzhou Normal University, Zhengzhou, People’s Republic of China
| | - Peiyuan Deng
- School of Life Sciences, Zhengzhou Normal University, Zhengzhou, People’s Republic of China
| | - Ping Chen
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| |
Collapse
|
43
|
Yang L, Wang Q, He L, Sun X. The critical role of tumor microbiome in cancer immunotherapy. Cancer Biol Ther 2024; 25:2301801. [PMID: 38241173 PMCID: PMC10802201 DOI: 10.1080/15384047.2024.2301801] [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: 10/24/2023] [Accepted: 01/01/2024] [Indexed: 01/21/2024] Open
Abstract
In recent years, the microbiome has shown an integral role in cancer immunotherapy and has become a prominent and widely studied topic. A full understanding of the interactions between the tumor microbiome and various immunotherapies offers opportunities for immunotherapy of cancer. This review scrutinizes the composition of the tumor microbiome, the mechanism of microbial immune regulation, the influence of tumor microorganisms on tumor metastasis, and the interaction between tumor microorganisms and immunotherapy. In addition, this review also summarizes the challenges and opportunities of immunotherapy through tumor microbes, as well as the prospects and directions for future related research. In conclusion, the potential of microbial immunotherapy to enhance treatment outcomes for cancer patients should not be underestimated. Through this review, it is hoped that more research on tumor microbial immunotherapy will be done to better solve the treatment problems of cancer patients.
Collapse
Affiliation(s)
- Liu Yang
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Lijuan He
- Department of Health Management Center, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Xingyu Sun
- Department of Gynecology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
44
|
Sepehr A, Miri ST, Aghamohammad S, Rahimirad N, Milani M, Pourshafie MR, Rohani M. Health benefits, antimicrobial activities, and potential applications of probiotics: A review. Medicine (Baltimore) 2024; 103:e32412. [PMID: 39969286 PMCID: PMC11688011 DOI: 10.1097/md.0000000000032412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 12/02/2022] [Indexed: 02/20/2025] Open
Abstract
Gut microbiota and its metabolic activities can influence the physiology and pathology of the human body. It is well established that alterations in the balance of living microbiota can contribute to various health problems, such as inflammatory bowel disease and autoimmune disorders. Probiotics administered in sufficient quantities as functional food ingredients provide health benefits to hosts. They help to maintain the stability and composition of the gut microbiota and provide resistance to infection by pathogens. The most important probiotic bacteria are Lactobacillus spp. and Bifidobacteria spp., which protect the intestine through various mechanisms such as the production of organic acids and bacteriocins. Scientific and clinical research has demonstrated that probiotics play a role in modulating immune response and preventing cancer and chronic inflammatory diseases, especially in the gastrointestinal tract. This article summarizes the potential health benefits, antimicrobial activities, and purposes for which probiotics can be used as functional foods to improve human health.
Collapse
Affiliation(s)
- Amin Sepehr
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | - Seyedeh Tina Miri
- Department of Microbiology, Islamic Azad University Science and Research Branch, Tehran, Iran
| | | | - Nazanin Rahimirad
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahnaz Milani
- Department of Microbiology, Islamic Azad University Science and Research Branch, Tehran, Iran
| | | | - Mahdi Rohani
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
45
|
Mechtersheimer D, Ding W, Xu X, Kim S, Sue C, Cao Y, Yang J. IMPACT: interpretable microbial phenotype analysis via microbial characteristic traits. Bioinformatics 2024; 41:btae702. [PMID: 39658259 PMCID: PMC11687948 DOI: 10.1093/bioinformatics/btae702] [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: 12/07/2023] [Revised: 08/23/2024] [Accepted: 12/09/2024] [Indexed: 12/12/2024] Open
Abstract
MOTIVATION The human gut microbiome, consisting of trillions of bacteria, significantly impacts health and disease. High-throughput profiling through the advancement of modern technology provides the potential to enhance our understanding of the link between the microbiome and complex disease outcomes. However, there remains an open challenge where current microbiome models lack interpretability of microbial features, limiting a deeper understanding of the role of the gut microbiome in disease. To address this, we present a framework that combines a feature engineering step to transform tabular abundance data to image format using functional microbial annotation databases, with a residual spatial attention transformer block architecture for phenotype classification. RESULTS Our model, IMPACT, delivers improved predictive accuracy performance across multiclass classification compared to similar methods. More importantly, our approach provides interpretable feature importance through image classification saliency methods. This enables the extraction of taxa markers (features) associated with a disease outcome and also their associated functional microbial traits and metabolites. AVAILABILITY AND IMPLEMENTATION IMPACT is available at https://github.com/SydneyBioX/IMPACT. We providedirect installation of IMPACT via pip.
Collapse
Affiliation(s)
- Daniel Mechtersheimer
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW 2006, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Wenze Ding
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW 2006, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Xiangnan Xu
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW 2006, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, NSW 2006, Australia
- Neuroscience Research Australia, Randwick, NSW 2031, Australia
| | - Sanghyun Kim
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW 2006, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Carolyn Sue
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
- Neuroscience Research Australia, Randwick, NSW 2031, Australia
| | - Yue Cao
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW 2006, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
- Laboratory of Data Discovery for Health Limited (D24H), New Territories, Hong Kong SAR, China
| | - Jean Yang
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW 2006, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
- Laboratory of Data Discovery for Health Limited (D24H), New Territories, Hong Kong SAR, China
| |
Collapse
|
46
|
Fedosova N, Cheremshenko N, Gogol S, Bezdieniezhnykh N, Tikhonov V, Voyeykova I, Konovalenko V, Chekhun V. THE INFLUENCE OF BIFIDOBACTERIUM ANIMALIS AND LECTIN OF B. SUBTILIS IMV B-7724 ON THE ANTITUMOR IMMUNE RESPONSE OF MICE WITH EHRLICH ADENOCARCINOMA. Exp Oncol 2024; 46:228-236. [PMID: 39704458 DOI: 10.15407/exp-oncology.2024.03.228] [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: 12/19/2024] [Indexed: 12/21/2024]
Abstract
AIM To investigate the effect of bacteria of the genus Bifidobacterium and the extracellular metabolite of B. subtilis IMV B-7724 on the antitumor immune response of mice with a model tumor. MATERIALS AND METHODS The study was conducted on Balb/c mice with transplanted solid Ehrlich adenocarcinoma (ACE). Starting from the 2nd day after the transplantation of tumor cells, the animals of the experimental groups were treated with lectin of B. subtilis IMV B-7724 (s/c, 1 mg/kg of weight), Bifidobacterium animalis (per os, 7 × 105 CFU/mouse) or their combination. The immunological studies were performed on the 21st and 28th days of tumor growth. The functional activity of natural killer cells (NK), cytotoxic T-lymphocytes (CTL), as well as the ability of lymphocytes from the peripheral lymph nodes (PLN) to transform into blast cells under the influence of T- (Con A) and B-cell (LPS) mitogens were determined. RESULTS Administration of probiotic components to the mice with ACE led to the activation of innate immune responses, that is, to a significant increase in the cytotoxic activity of NK, especially in the case of their combined use. The NK cytotoxicity index was higher than that in the non-treated ACE-bearing mice and the intact control by 3.7 and 2.1 times, respectively (p < 0.05). Similarly, the highest specific cytotoxic activity of spleen lymphocytes was observed upon the combined use of the microbial preparations: the CTL cytotoxicity index was nearly 2.5-fold higher than in the non-treated ACE-bearing mice. The data on the ability of PLN lymphocytes to transform into blast cells under the influence of Con A and LPS indicated the preservation of the functional activity of lymphocytes in the animals of the experimental groups during ACE growth. CONCLUSION Both B. animalis and lectin of B. subtilis IMV B-7724 have a significant influence on the effectors of the natural and adaptive immunity of mice with ACE. Their combined use was found to be the most effective.
Collapse
Affiliation(s)
- N Fedosova
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology of the NAS of Ukraine, Kyiv, Ukraine
| | - N Cheremshenko
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology of the NAS of Ukraine, Kyiv, Ukraine
| | - S Gogol
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology of the NAS of Ukraine, Kyiv, Ukraine
| | - N Bezdieniezhnykh
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology of the NAS of Ukraine, Kyiv, Ukraine
| | - V Tikhonov
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology of the NAS of Ukraine, Kyiv, Ukraine
| | - I Voyeykova
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology of the NAS of Ukraine, Kyiv, Ukraine
| | - V Konovalenko
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology of the NAS of Ukraine, Kyiv, Ukraine
| | - V Chekhun
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology of the NAS of Ukraine, Kyiv, Ukraine
| |
Collapse
|
47
|
Tsuchida S, Umemura H, Iizuka K, Yamamoto H, Shimazaki I, Shikata E, Nakayama T. Recent findings on metabolomics and the microbiome of oral bacteria involved in dental caries and periodontal disease. World J Microbiol Biotechnol 2024; 41:11. [PMID: 39690257 DOI: 10.1007/s11274-024-04224-3] [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: 02/07/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024]
Abstract
Periodontal disease is characterized by bacterial toxins within the oral biofilm surrounding the teeth, leading to gingivitis and the gradual dissolution of the alveolar bone, which supports the teeth. Notably, symptoms in the early stages of the disease are often absent. Similarly, dental caries occurs when oral bacteria metabolize dietary sugars, producing acids that dissolve tooth enamel and dentin. These bacteria are commonly present in the oral cavity of most individuals. Metabolomics, a relatively recent addition to the "omics" research landscape, involves the comprehensive analysis of metabolites in vivo to elucidate pathological mechanisms and accelerate drug discovery. Meanwhile, the term "microbiome" refers to the collection of microorganisms within a specific environmental niche or their collective genomes. The human microbiome plays a critical role in health and disease, influencing a wide array of physiological and pathological processes. Recent advances in microbiome research have identified numerous bacteria implicated in dental caries and periodontal disease. Additionally, studies have uncovered various pathogenic factors associated with these microorganisms. This review focuses on recent findings in metabolomics and the microbiome, specifically targeting oral bacteria linked to dental caries and periodontal disease. We acknowledge the limitation of relying exclusively on the MEDLINE database via PubMed, while excluding other sources such as gray literature, conference proceedings, and clinical practice guidelines.
Collapse
Affiliation(s)
- Sachio Tsuchida
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Hiroshi Umemura
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Kazuhide Iizuka
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Haruka Yamamoto
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Isamu Shimazaki
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Elisa Shikata
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Tomohiro Nakayama
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan.
| |
Collapse
|
48
|
Roberts KD, Ahmed S, San Valentin E, Di Martino L, McCormick TS, Ghannoum MA. Immunomodulatory Properties of Multi-Strain Postbiotics on Human CD14 + Monocytes. Life (Basel) 2024; 14:1673. [PMID: 39768380 PMCID: PMC11728152 DOI: 10.3390/life14121673] [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: 11/08/2024] [Revised: 12/06/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
The ability of probiotics, comprising live microbiota, to modulate the composition of intestinal microbiomes has been connected to modulation of the central nervous system (Gut-Brain axis), neuroendocrine system (Gut-Skin axis), and immune response (Gut-Immune axis). Less information is known regarding the ability of postbiotics (cell wall components and secreted metabolites derived from live organisms) to regulate host immunity. In the present study, we tested postbiotics comprising single strains of bacteria and yeast (Lactobacillus acidophilus 16axg, Lacticaseibacillus rhamnosus 18fx, Saccharomyces cerevisiae var. boulardii 16mxg) as well as combinations of multiple strains for their ability to stimulate cytokine production by human CD14+ monocytes. We quantified cytokine gene and protein expression levels in monocytes following stimulation with postbiotics. Both heat-killed L. acidophilus and L. rhamnosus stimulated naïve monocytes without significant differences between them. Heat-killed S. boulardii stimulated less cytokine production compared to postbiotic bacteria at the same concentration. Interestingly, the addition of heat-killed yeast to heat-killed L. acidophilus and L. rhamnosus resulted in an enhancement of immune stimulation. Thus, heat-killed postbiotics have immune-modulating potential, particularly when bacteria and yeast are combined. This approach may hold promise for developing targeted interventions that can be fine-tuned to modulate host immune response with beneficial health impact.
Collapse
Affiliation(s)
- Kyle D. Roberts
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Sadia Ahmed
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Erin San Valentin
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Luca Di Martino
- Case Digestive Health Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Thomas S. McCormick
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Mahmoud A. Ghannoum
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
49
|
Korsirikoon C, Techaniyom P, Kettawan A, Rungruang T, Metheetrairut C, Prombutara P, Kettawan AK. Cold-pressed extraction of perilla seed oil enriched with alpha-linolenic acid mitigates tumour progression and restores gut microbial homeostasis in the AOM/DSS mice model of colitis-associated colorectal cancer. PLoS One 2024; 19:e0315172. [PMID: 39652552 PMCID: PMC11627366 DOI: 10.1371/journal.pone.0315172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024] Open
Abstract
The present investigation explores into the influence of dietary nutrients, particularly alpha-linolenic acid (ALA), a plant-derived omega-3 fatty acid abundant in perilla seed oil (PSO), on the development of colitis-associated colorectal cancer (CRC). The study employs a mouse model to scrutinize the effects of ALA-rich PSO in the context of inflammation-driven CRC. Perilla seeds were subjected to oil extraction, and the nutritional composition of the obtained oil was analysed. Male ICR mice, initiated at four weeks of age, were subjected to diets comprising 5%, 10%, or 20% PSO, 10% fish oil, or 5% soybean oil. All groups, with the exception of the control group (5% soybean oil), underwent induction with azoxymethane (AOM) and dextran sulphate sodium (DSS) to instigate CRC. Disease development, colon samples, preneoplastic lesions, dysplasia, and biomarkers were meticulously evaluated. Furthermore, gut microbiota composition was elucidated through 16S rRNA sequencing. The analysis revealed that PSO contained 61.32% ALA and 783.90 mg/kg tocopherols. Mice subjected to diets comprising 5% soybean or 10% fish oil exhibited higher tumour incidence, burden, multiplicity, and aberrant crypt counts. Remarkably, these parameters were significantly reduced in mice fed a 5% PSO diet. Additionally, 5% PSO-fed mice displayed reduced proliferative and pro-inflammatory markers in colon tissues, coupled with an alleviation of AOM/DSS-induced gut dysbiosis. Notably, PSO demonstrated inhibitory effects on colitis-associated CRC in the AOM/DSS mice model, achieved through the suppression of proliferative and pro-inflammatory protein levels, and mitigation of gut dysbiosis, with discernible efficacy observed at a 5% dietary concentration.
Collapse
Affiliation(s)
- Chawin Korsirikoon
- Doctor of Philosophy Program in Nutrition, Faculty of Medicine Ramathibodi Hospital and Institute of Nutrition, Mahidol University, Bangkok, Thailand
| | - Peerapa Techaniyom
- Doctor of Philosophy Program in Nutrition, Faculty of Medicine Ramathibodi Hospital and Institute of Nutrition, Mahidol University, Bangkok, Thailand
| | | | - Thanaporn Rungruang
- Department of Anatomy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chanatip Metheetrairut
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pinidphon Prombutara
- OMICS Sciences and Bioinformatics Center, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | | |
Collapse
|
50
|
Hashimoto A, Hashimoto S. Plasticity and Tumor Microenvironment in Pancreatic Cancer: Genetic, Metabolic, and Immune Perspectives. Cancers (Basel) 2024; 16:4094. [PMID: 39682280 DOI: 10.3390/cancers16234094] [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: 11/09/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer has long been believed to be a genetic disease caused by the accumulation of mutations in key genes involved in cellular processes. However, recent advances in sequencing technology have demonstrated that cells with cancer driver mutations are also present in normal tissues in response to aging, environmental damage, and chronic inflammation, suggesting that not only intrinsic factors within cancer cells, but also environmental alterations are important key factors in cancer development and progression. Pancreatic cancer tissue is mostly comprised of stromal cells and immune cells. The desmoplasmic microenvironment characteristic of pancreatic cancer is hypoxic and hypotrophic. Pancreatic cancer cells may adapt to this environment by rewiring their metabolism through epigenomic changes, enhancing intrinsic plasticity, creating an acidic and immunosuppressive tumor microenvironment, and inducing noncancerous cells to become tumor-promoting. In addition, pancreatic cancer has often metastasized to local and distant sites by the time of diagnosis, suggesting that a similar mechanism is operating from the precancerous stage. Here, we review key recent findings on how pancreatic cancers acquire plasticity, undergo metabolic reprogramming, and promote immunosuppressive microenvironment formation during their evolution. Furthermore, we present the following two signaling pathways that we have identified: one based on the small G-protein ARF6 driven by KRAS/TP53 mutations, and the other based on the RNA-binding protein Arid5a mediated by inflammatory cytokines, which promote both metabolic reprogramming and immune evasion in pancreatic cancer. Finally, the striking diversity among pancreatic cancers in the relative importance of mutational burden and the tumor microenvironment, their clinical relevance, and the potential for novel therapeutic strategies will be discussed.
Collapse
Affiliation(s)
- Ari Hashimoto
- Department of Molecular Biology, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Shigeru Hashimoto
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0818, Japan
| |
Collapse
|