101
|
Liu X, Li X, Li H, Guan B, Jiang Y, Zheng C, Kong D. Annexin A1: a key regulator of T cell function and bone marrow adiposity in aplastic anaemia. J Physiol 2024; 602:6125-6152. [PMID: 39373986 DOI: 10.1113/jp286148] [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: 12/15/2023] [Accepted: 07/31/2024] [Indexed: 10/08/2024] Open
Abstract
This study investigates the role of Annexin A1 (ANXA1) in regulating T cell function and its implications in bone marrow adiposity in aplastic anaemia (AA). Utilizing single-cell sequencing analysis, we compared bone marrow tissues from AA patients and healthy individuals, focusing on T cell subgroups and their impact on bone marrow pathology. Our findings reveal a significant activation of CD8+ T cells in AA, driven by reduced ANXA1 expression. This heightened T cell activity promotes adipogenesis in bone marrow-derived mesenchymal stem cells via IFN-γ secretion. Overexpression of ANXA1 was found to suppress this process, suggesting its therapeutic potential in AA treatment. The study highlights ANXA1 as a crucial regulator in the AA-associated immune microenvironment and bone marrow adiposity. KEY POINTS: This study found that ANXA1 is significantly downregulated in AA and provides detailed insights into its critical role in the disease. The study demonstrates the excessive activation of CD8+ T cells in the progression of AA. The research shows that the overexpression of ANXA1 can effectively inhibit the activation of CD8+ T cells. The study confirms that overexpression of ANXA1 reduces the secretion of the cytokine IFN-γ, decreases adipogenesis in bone marrow-derived mesenchymal stem cells and may improve AA symptoms. This research provides new molecular targets for the treatment of AA.
Collapse
Affiliation(s)
- Xia Liu
- Department of Respiratory Intervention, Children's Hospital Affiliated to Shandong University, Jinan, China
| | - Xiaomei Li
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- JiNan Key Laboratory of Basic and Clinical Translational Research in Radiobiology, Jinan, China
| | - Hui Li
- Department of Hematology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bingxin Guan
- Department of Pathology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yang Jiang
- Institute of Biotherapy for Hematological Malignancies, Shandong University, Jinan, China
- Shandong University-Karolinska Institute Collaborative Laboratory for Stem Cell Research, Shandong University, Jinan, China
| | - Chengyun Zheng
- Department of Hematology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute of Biotherapy for Hematological Malignancies, Shandong University, Jinan, China
- Shandong University-Karolinska Institute Collaborative Laboratory for Stem Cell Research, Shandong University, Jinan, China
| | - Dexiao Kong
- Department of Hematology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute of Biotherapy for Hematological Malignancies, Shandong University, Jinan, China
- Shandong University-Karolinska Institute Collaborative Laboratory for Stem Cell Research, Shandong University, Jinan, China
| |
Collapse
|
102
|
Duan Y, Dai J, Lu Y, Qiao H, Liu N. Disentangling the molecular mystery of tumour-microbiota interactions: Microbial metabolites. Clin Transl Med 2024; 14:e70093. [PMID: 39568157 PMCID: PMC11578933 DOI: 10.1002/ctm2.70093] [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: 06/24/2024] [Revised: 09/27/2024] [Accepted: 11/02/2024] [Indexed: 11/22/2024] Open
Abstract
The profound impact of the microbiota on the initiation and progression of cancer has been a focus of attention. In recent years, many studies have shown that microbial metabolites serve as key hubs that connect the microbiome and cancer progression, but the underlying molecular mechanisms have not been fully elucidated. Multiple mechanisms that influence tumour development and therapy resistance, including disrupting cellular signalling pathways, triggering oxidative stress, inducing metabolic reprogramming and reshaping tumour immune microenvironment, are reviewed. Focusing on recent advancements in this field, this review also summarises the methodological framework of studies regarding microbial metabolites. In this review, we outline the current state of research on tumour-associated microbial metabolites and describe the challenges in future scientific research and clinical applications. KEY POINTS: Metabolites derived from both gut and intratumoural microbiota play important roles in cancer initiation and progression. The dual roles of microbial metabolites pose an obstacle for clinical translations. Absolute quantification and tracing techniques of microbial metabolites are essential for addressing the gaps in studies on microbial metabolites. Integrating microbial metabolomics with multi-omics transcends current research paradigms.
Collapse
Affiliation(s)
- Yu‐Fei Duan
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouPR China
| | - Jia‐Hao Dai
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouPR China
| | - Ying‐Qi Lu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouPR China
| | - Han Qiao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouPR China
| | - Na Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouPR China
| |
Collapse
|
103
|
Khan M, Dong Y, Ullah R, Li M, Huang Q, Hu Y, Yang L, Luo Z. Recent Advances in Bacterium-Based Therapeutic Modalities for Melanoma Treatment. Adv Healthc Mater 2024; 13:e2401076. [PMID: 39375965 DOI: 10.1002/adhm.202401076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/16/2024] [Indexed: 10/09/2024]
Abstract
Melanoma is one of the most severe skin cancer indications with rapid progression and a high risk of metastasis. However, despite the accumulated advances in melanoma treatment including adjuvant radiation, chemotherapy, and immunotherapy, the overall melanoma treatment efficacy in the clinics is still not satisfactory. Interestingly, bacterial therapeutics have demonstrated unique properties for tumor-related therapeutic applications, such as tumor-targeted motility, tailorable cytotoxicity, and immunomodulatory capacity of the tumor microenvironment, which have emerged as a promising platform for melanoma therapy. Indeed, the recent advances in genetic engineering and nanotechnologies have boosted the application potential of bacterium-based therapeutics for treating melanoma by further enhancing their tumor-homing, cell-killing, drug delivery, and immunostimulatory capacities. This review provides a comprehensive summary of the state-of-the-art bacterium-based anti-melanoma modalities, which are categorized according to their unique functional merits, including tumor-specific cytotoxins, tumor-targeted drug delivery platforms, and immune-stimulatory agents. Furthermore, a perspective is provided discussing the potential challenges and breakthroughs in this area. The insights in this review may facilitate the development of more advanced bacterium-based therapeutic modalities for improved melanoma treatment efficacy.
Collapse
Affiliation(s)
- Mubassir Khan
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Yilong Dong
- Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325016, P. R. China
| | - Razi Ullah
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Lab for Vascular Implants College of Bioengineering Chongqing University, Chongqing, 400030, P. R. China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing, 400044, P. R. China
| | - Qiping Huang
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing, 400044, P. R. China
| |
Collapse
|
104
|
Ma C, Zhang S, Renaud SJ, Zhang Q, Qi H, Zhou H, Jin Y, Yu H, Xu Y, Huang H, Hong Y, Li H, Liao Q, Ding F, Qin M, Wang P, Xie Z. Structural elucidation of a capsular polysaccharide from Bacteroides uniformis and its ameliorative impact on DSS-induced colitis in mice. Int J Biol Macromol 2024; 279:135119. [PMID: 39208897 DOI: 10.1016/j.ijbiomac.2024.135119] [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: 03/29/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Capsular polysaccharides derived from Bacteroides species have emerged as potential mitigators of intestinal inflammation in murine models. However, research on capsular polysaccharides from B. uniformis, a Bacteroides species with reduced abundance in colons of patients with ulcerative colitis, remains scarce. In this study, we extracted a neutral polysaccharide component from B. uniformis ATCC8492, termed BUCPS1B, using ultrasonic disruption, ethanol precipitation, and anion exchange chromatography. Structural characterization revealed BUCPS1B as a water-soluble polysaccharide with an α-1,4-glucan main chain adorned with minor substituent sugar residues. BUCPS1B alleviated intestinal inflammation in a mouse model of colitis and induced polarization of macrophages into M2-type. Furthermore, BUCPS1B modulated the gut microbiota composition, increased the abundance of the probiotic Akkermansia muciniphila and altered the gut metabolic profile to promote phenylalanine and short chain fatty acids metabolism. BUCPS1B is therefore a promising candidate to prevent inflammation and augment intestinal health.
Collapse
Affiliation(s)
- Chong Ma
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510006, PR China
| | - Shaobao Zhang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510006, PR China
| | - Stephen James Renaud
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario, Canada
| | - Qian Zhang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510006, PR China
| | - Huiyuan Qi
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510006, PR China
| | - Haiyun Zhou
- Instrumental Analysis & Research Center, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Yibao Jin
- National Medical Products Administration, Shenzhen Institute for Drug Control, Shenzhen, PR China
| | - Hansheng Yu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510006, PR China
| | - Yaning Xu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510006, PR China
| | - Houshuang Huang
- National Medical Products Administration, Shenzhen Institute for Drug Control, Shenzhen, PR China
| | - Yanjun Hong
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510006, PR China
| | - Hao Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510006, PR China
| | - Qiongfeng Liao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Feiqing Ding
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510006, PR China
| | - Meirong Qin
- National Medical Products Administration, Shenzhen Institute for Drug Control, Shenzhen, PR China
| | - Ping Wang
- National Medical Products Administration, Shenzhen Institute for Drug Control, Shenzhen, PR China.
| | - Zhiyong Xie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510006, PR China.
| |
Collapse
|
105
|
Couture G, Cheang SE, Suarez C, Chen Y, Bacalzo NP, Jiang J, Weng CYC, Stacy A, Castillo JJ, Delannoy-Bruno O, Webber DM, Barratt MJ, Gordon JI, Mills DA, German JB, Fukagawa NK, Lebrilla CB. A multi-glycomic platform for the analysis of food carbohydrates. Nat Protoc 2024; 19:3321-3359. [PMID: 39026121 DOI: 10.1038/s41596-024-01017-8] [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: 12/12/2022] [Accepted: 04/30/2024] [Indexed: 07/20/2024]
Abstract
Carbohydrates comprise the largest fraction of most diets and exert a profound impact on health. Components such as simple sugars and starch supply energy, while indigestible components, deemed dietary fiber, reach the colon to provide food for the tens of trillions of microbes that make up the gut microbiota. The interactions between dietary carbohydrates, our gastrointestinal tracts, the gut microbiome and host health are dictated by their structures. However, current methods for analysis of food glycans lack the sensitivity, specificity and throughput needed to quantify and elucidate these myriad structures. This protocol describes a multi-glycomic approach to food carbohydrate analysis in which the analyte might be any food item or biological material such as fecal and cecal samples. The carbohydrates are extracted by ethanol precipitation, and the resulting samples are subjected to rapid-throughput liquid chromatography (LC)-tandem mass spectrometry (LC-MS/MS) methods. Quantitative analyses of monosaccharides, glycosidic linkages, polysaccharides and alcohol-soluble carbohydrates are performed in 96-well plates at the milligram scale to reduce the biomass of sample required and enhance throughput. Detailed stepwise processes for sample preparation, LC-MS/MS and data analysis are provided. We illustrate the application of the protocol to a diverse set of foods as well as different apple cultivars and various fermented foods. Furthermore, we show the utility of these methods in elucidating glycan-microbe interactions in germ-free and colonized mice. These methods provide a framework for elucidating relationships between dietary fiber, the gut microbiome and human physiology. These structures will further guide nutritional and clinical feeding studies that enhance our understanding of the role of diet in nutrition and health.
Collapse
Affiliation(s)
- Garret Couture
- Department of Chemistry, University of California, Davis, Davis, CA, USA
- Foods for Health Institute, University of California, Davis, Davis, CA, USA
| | - Shawn Ehlers Cheang
- Department of Chemistry, University of California, Davis, Davis, CA, USA
- Foods for Health Institute, University of California, Davis, Davis, CA, USA
| | - Christopher Suarez
- Department of Chemistry, University of California, Davis, Davis, CA, USA
- Foods for Health Institute, University of California, Davis, Davis, CA, USA
| | - Ye Chen
- Department of Chemistry, University of California, Davis, Davis, CA, USA
- Foods for Health Institute, University of California, Davis, Davis, CA, USA
| | - Nikita P Bacalzo
- Department of Chemistry, University of California, Davis, Davis, CA, USA
- Foods for Health Institute, University of California, Davis, Davis, CA, USA
| | - Jiani Jiang
- Department of Chemistry, University of California, Davis, Davis, CA, USA
- Foods for Health Institute, University of California, Davis, Davis, CA, USA
| | - Cheng-Yu Charlie Weng
- Department of Chemistry, University of California, Davis, Davis, CA, USA
- Foods for Health Institute, University of California, Davis, Davis, CA, USA
| | - Aaron Stacy
- Department of Chemistry, University of California, Davis, Davis, CA, USA
- Foods for Health Institute, University of California, Davis, Davis, CA, USA
| | - Juan J Castillo
- Department of Chemistry, University of California, Davis, Davis, CA, USA
- Foods for Health Institute, University of California, Davis, Davis, CA, USA
| | - Omar Delannoy-Bruno
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St Louis, MO, USA
| | - Daniel M Webber
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Michael J Barratt
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Jeffrey I Gordon
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - David A Mills
- Foods for Health Institute, University of California, Davis, Davis, CA, USA
- Department of Food Science and Technology, University of California, Davis, Davis, CA, USA
- Department of Viticulture and Enology, University of California, Davis, Davis, CA, USA
| | - J Bruce German
- Foods for Health Institute, University of California, Davis, Davis, CA, USA
- Department of Food Science and Technology, University of California, Davis, Davis, CA, USA
| | - Naomi K Fukagawa
- USDA Agricultural Research Service, Beltsville Human Nutrition Research Center, Beltsville, MD, USA
| | - Carlito B Lebrilla
- Department of Chemistry, University of California, Davis, Davis, CA, USA.
- Foods for Health Institute, University of California, Davis, Davis, CA, USA.
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
106
|
Fu Y, Guo X, Sun L, Cui T, Wang J, Liu Y, Liu L. Exploring the Interplay of Diet, Obesity, Immune Function, and Cancer. Cancer Discov 2024; 14:2047-2050. [PMID: 39485246 DOI: 10.1158/2159-8290.cd-24-0834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/29/2024] [Accepted: 09/03/2024] [Indexed: 11/03/2024]
Abstract
This commentary provides an in-depth exploration of the intricate relationships among diet, obesity, immune function, and cancer, highlighting the potential role of dietary interventions as complementary therapies in cancer treatment. Multiple analyses underscore the importance of personalized dietary strategies in cancer management and identify opportunities for further research in this evolving field.
Collapse
Affiliation(s)
- Yumin Fu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, China
| | - Xinyu Guo
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, China
| | - Linmao Sun
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, China
| | - Tianming Cui
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, China
| | - Jiabei Wang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, China
| | - Yao Liu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, China
| | - Lianxin Liu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, China
| |
Collapse
|
107
|
Dreisbach C, Nansel T, Peddada S, Nicholson W, Siega-Riz AM. Dietary Sugar and Saturated Fat Consumption Associated with the Gastrointestinal Microbiome during Pregnancy. J Nutr 2024; 154:3246-3254. [PMID: 39307280 PMCID: PMC11600114 DOI: 10.1016/j.tjnut.2024.09.016] [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: 10/06/2023] [Revised: 07/29/2024] [Accepted: 09/10/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Growing evidence supports changes in the gastrointestinal microbiome over the course of pregnancy may have an impact on the short- and long-term health of both the mother and the child. OBJECTIVE Our objective was to explore the association of diet quality, as measured by the Healthy Eating Index (HEI), with the composition and gene ontology (GO) representation of microbial function in the maternal gastrointestinal microbiome during pregnancy. METHODS We conducted a retrospective, observational analysis of n = 185 pregnant participants in the Pregnancy Eating Attributes Study. Maternal dietary intake was assessed in the first trimester using the automated self-administered 24-h recall method, from which the HEI 2015 was calculated. Rectal swabs were obtained in the second trimester and sequenced using the NovaSeq 6000 system shotgun platform. We used unsupervised clustering to identify microbial enterotypes representative of maternal taxa and GO functional term composition. Multivariable linear models were used to identify associations between taxa, functional terms, and food components while controlling for relevant covariates. Multinomial regression was then used to predict enterotype membership based on a participant's HEI food component score. RESULTS Those in the high diet quality tertile had a lower early pregnancy BMI [mean (M) = 23.48 kg/m2, SD = 3.38] compared with the middle (M = 27.35, SD = 6.01) and low (M = 27.49, SD = 6.99) diet quality tertiles (P < 0.01). There were no statistically significant associations between the HEI components or the total HEI score and the 4 alpha diversity measures. Differences in taxa and GO term enterotypes were found in participants with, but not limited to, a higher saturated fat component score (β = 1.35, P = 0.01), added sugar HEI component (β = 0.07, P < 0.001), and higher total dairy score (β = 1.58, P = 0.01). CONCLUSIONS Specific dietary components are associated with microbial composition and function in the second trimester of pregnancy. These findings provide a foundation for future testable hypotheses.
Collapse
Affiliation(s)
- Caitlin Dreisbach
- School of Nursing, University of Rochester, Rochester, NY, United States; Goergen Institute for Data Science, University of Rochester, Rochester, NY, United States.
| | - Tonja Nansel
- Social and Behavioral Sciences Branch, Division of Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, United States
| | - Shyamal Peddada
- Biostatistics & Computational Biology Branch, National Institute for Environmental Health Sciences, Bethesda, MD, United States
| | - Wanda Nicholson
- Milken Institute School of Public Health, George Washington University, Washington, DC, United States
| | - Anna Maria Siega-Riz
- School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| |
Collapse
|
108
|
Xu C, Guo J, Chang B, Zhang Y, Tan Z, Tian Z, Duan X, Ma J, Jiang Z, Hou J. Design of probiotic delivery systems and their therapeutic effects on targeted tissues. J Control Release 2024; 375:20-46. [PMID: 39214316 DOI: 10.1016/j.jconrel.2024.08.037] [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: 03/05/2024] [Revised: 08/14/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
The microbiota at different sites in the body is closely related to disease. The intake of probiotics is an effective strategy to alleviate diseases and be adjuvant in their treatment. However, probiotics may suffer from harsh environments and colonization resistance, making it difficult to maintain a sufficient number of live probiotics to reach the target sites and exert their original probiotic effects. Encapsulation of probiotics is an effective strategy. Therefore, probiotic delivery systems, as effective methods, have been continuously developed and innovated to ensure that probiotics are effectively delivered to the targeted site. In this review, initially, the design of probiotic delivery systems is reviewed from four aspects: probiotic characteristics, processing technologies, cell-derived wall materials, and interactions between wall materials. Subsequently, the review focuses on the effects of probiotic delivery systems that target four main microbial colonization sites: the oral cavity, skin, intestine, and vagina, as well as disease sites such as tumors. Finally, this review also discusses the safety concerns of probiotic delivery systems in the treatment of disease and the challenges and limitations of implementing this method in clinical studies. It is necessary to conduct more clinical studies to evaluate the effectiveness of different probiotic delivery systems in the treatment of diseases.
Collapse
Affiliation(s)
- Cong Xu
- College of Food Science and Engineering, Guiyang University, Guiyang 550005, China; Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, China; Heilongjiang Green Food Science Research Institute, Harbin 150028, China
| | - Jiahui Guo
- Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, China
| | - Baoyue Chang
- Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, China
| | - Yiming Zhang
- Department of Psychiatry and Mental Health, Dalian Medical University, Dalian 116044, China
| | - Zhongmei Tan
- Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, China
| | - Zihao Tian
- Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, China
| | - Xiaolei Duan
- Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, China
| | - Jiage Ma
- Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, China; Heilongjiang Green Food Science Research Institute, Harbin 150028, China
| | - Zhanmei Jiang
- Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, China.
| | - Juncai Hou
- College of Food Science and Engineering, Guiyang University, Guiyang 550005, China; Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, China; Heilongjiang Green Food Science Research Institute, Harbin 150028, China.
| |
Collapse
|
109
|
Bohm MS, Ramesh AV, Pierre JF, Cook KL, Murphy EA, Makowski L. Fecal microbial transplants as investigative tools in cancer. Am J Physiol Gastrointest Liver Physiol 2024; 327:G711-G726. [PMID: 39301964 PMCID: PMC11559651 DOI: 10.1152/ajpgi.00171.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/22/2024]
Abstract
The gut microbiome plays a critical role in the development, progression, and treatment of cancer. As interest in microbiome-immune-cancer interactions expands, the prevalence of fecal microbial transplant (FMT) models has increased proportionally. However, current literature does not provide adequate details or consistent approaches to allow for necessary rigor and experimental reproducibility. In this review, we evaluate key studies using FMT to investigate the relationship between the gut microbiome and various types of cancer. In addition, we will discuss the common pitfalls of these experiments and methods for improved standardization and validation as the field uses FMT with greater frequency. Finally, this review focuses on the impacts of the gut and extraintestinal microbes, prebiotics, probiotics, and postbiotics in cancer risk and response to therapy across a variety of tumor types.NEW & NOTEWORTHY The microbiome impacts the onset, progression, and therapy response of certain types of cancer. Fecal microbial transplants (FMTs) are an increasingly prevalent tool to test these mechanisms that require standardization by the field.
Collapse
Affiliation(s)
- Margaret S Bohm
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Arvind V Ramesh
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Joseph F Pierre
- Department of Nutritional Sciences, College of Agriculture and Life Science, The University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Katherine L Cook
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - E Angela Murphy
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Liza Makowski
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| |
Collapse
|
110
|
Jurlander RS, Guldbrandt LM, Holmstroem RB, Madsen K, Donia M, Haslund CA, Schmidt H, Bastholt L, Ruhlmann CH, Svane IM, Ellebaek E. Immune-related adverse events in a nationwide cohort of real-world melanoma patients treated with adjuvant anti-PD1 - Seasonal variation and association with outcome. Eur J Cancer 2024; 212:115053. [PMID: 39405648 DOI: 10.1016/j.ejca.2024.115053] [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: 06/18/2024] [Revised: 09/11/2024] [Accepted: 09/23/2024] [Indexed: 11/03/2024]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) carry the risk of immune-related adverse events (irAEs), a significant concern as therapy has transitioned to the adjuvant setting. Balancing therapeutic benefits against potential risks is crucial, necessitating real-world data from an unselected patient population in addition to clinical trial data to ensure optimal clinical decision-making. METHODS This nationwide real-world study assessed irAEs in patients receiving adjuvant anti-PD1 therapy, primarily nivolumab, for resected stage III-IV melanoma between 2018-2022. Data were retrieved from two national databases: the IMMUNOTOX database and the Danish Metastatic Melanoma Database (DAMMED). IrAEs were sub-grouped according to organ systems graded using CTCAE ver. 5.0 ranging from mild toxicities (grade 1-2) to severe (grade 3-4) and fatal (grade 5). RESULTS Among 792 included patients, (55 % male, median age 62 years (range 16-88)), 697 patients (88 %) experienced an irAE. Severe irAEs occurred in 116 patients (15 %) and five (0.6 %) died due to toxicity. A landmark analysis showed that patients who experienced at least one irAE before the 1st evaluation at 90 days had an increased progression free survival (PFS) (p = 0.032) and overall survival (OS) (p = 0.0071). Additionally, a seasonal pattern was noted with higher incidence of irAEs during summer. CONCLUSION The prevalence of irAEs in real-world patients is comparable to the observed risk in clinical trials. Patients experiencing irAEs demonstrate a lower risk of melanoma relapse. Further, gender, age and seasonal variation may impact the incidence of irAEs.
Collapse
Affiliation(s)
- Rebecca Schou Jurlander
- Center for Cancer Immunotherapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Denmark
| | | | - Rikke B Holmstroem
- Center for Cancer Immunotherapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Denmark
| | - Kasper Madsen
- Center for Cancer Immunotherapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Denmark
| | - Marco Donia
- Center for Cancer Immunotherapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Denmark
| | | | - Henrik Schmidt
- Department of Oncology, Aarhus University Hospital, Denmark
| | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Denmark
| | | | - Inge Marie Svane
- Center for Cancer Immunotherapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Denmark.
| | - Eva Ellebaek
- Center for Cancer Immunotherapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Denmark.
| |
Collapse
|
111
|
Scales TQ, Smith B, Blanchard LM, Wixom N, Tuttle ET, Altman BJ, Peppone LJ, Munger J, Campbell TM, Campbell EK, Harris IS. A whole food, plant-based diet reduces amino acid levels in patients with metastatic breast cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.09.24315165. [PMID: 39417128 PMCID: PMC11483017 DOI: 10.1101/2024.10.09.24315165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Background Amino acids are critical to tumor survival. Tumors can acquire amino acids from the surrounding microenvironment, including the serum. Limiting dietary amino acids is suggested to influence their serum levels. Further, a plant-based diet is reported to contain fewer amino acids than an animal-based diet. The extent to which a plant-based diet lowers the serum levels of amino acids in patients with cancer is unclear. Methods Patients with metastatic breast cancer (n=17) were enrolled in a clinical trial with an ad libitum whole food, plant-based diet for 8 weeks without calorie or portion restriction. Dietary changes by participants were monitored using a three-day food record. Serum was collected from participants at baseline and 8 weeks. Food records and serum were analyzed for metabolic changes. Results We found that a whole food, plant-based diet resulted in a lower intake of calories, fat, and amino acids and higher levels of fiber. Additionally, body weight, serum insulin, and IGF were reduced in participants. The diet contained lower levels of essential and non-essential amino acids, except for arginine (glutamine and asparagine were not measured). Importantly, the lowered dietary intake of amino acids translated to reduced serum levels of amino acids in participants (5/9 essential amino acids; 4/11 non-essential amino acids). Conclusions These findings provide a tractable approach to limiting amino acid levels in persons with cancer. This data lays a foundation for studying the relationship between amino acids in patients and tumor progression. Further, a whole-food, plant-based diet has the potential to synergize with cancer therapies that exploit metabolic vulnerabilities. Trial Registration The clinical trial was registered with ClinicalTrials.gov identifier NCT03045289 on 2017-02-07.
Collapse
Affiliation(s)
- TashJaé Q. Scales
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Bradley Smith
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Lisa M. Blanchard
- Department of Family Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, USA 14642
| | - Nellie Wixom
- Clinical Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Emily T. Tuttle
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Brian J. Altman
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Luke J. Peppone
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Department of Surgery, Cancer Control, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Joshua Munger
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Thomas M. Campbell
- Department of Family Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, USA 14642
| | - Erin K. Campbell
- Department of Public Health Sciences, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, USA,14642
| | - Isaac S. Harris
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| |
Collapse
|
112
|
Zhang J, Gan H, Duan X, Li G. Targeting the Intestinal Microbiota: A Novel Direction in the Treatment of Inflammatory Bowel Disease. Biomedicines 2024; 12:2340. [PMID: 39457652 PMCID: PMC11504502 DOI: 10.3390/biomedicines12102340] [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: 09/22/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Over the past decade, there has been a rapid increase in the incidence of inflammatory bowel disease. It has been suggested that multifactorial interactions of environmental factors, genetic factors, immune response and intestinal microbiota are involved in the pathogenesis of inflammatory bowel disease. It is widely recognized that the intestinal microbiota are essential for human metabolism, the immune system and pathogen resistance, and are integral to human health. Therefore, the dysbiosis of the microbiota is a critical step leading to intestinal mucosal damage and a key factor in the pathogenesis of inflammatory bowel disease. Regulating the microbiota through interventions such as enteral nutrition, fecal microbiota transplantation, and probiotic supplementation has the potential to prevent or even reverse intestinal dysbiosis, opening up new perspectives for the treatment of inflammatory bowel disease.
Collapse
Affiliation(s)
| | | | - Xiaoyan Duan
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Yangpu District, Shanghai 200092, China; (J.Z.); (H.G.)
| | - Guangming Li
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Yangpu District, Shanghai 200092, China; (J.Z.); (H.G.)
| |
Collapse
|
113
|
Liu C, Fu L, Wang Y, Yang W. Influence of the gut microbiota on immune cell interactions and cancer treatment. J Transl Med 2024; 22:939. [PMID: 39407240 PMCID: PMC11476117 DOI: 10.1186/s12967-024-05709-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
The tumour microenvironment represents a novel frontier in oncological research. Over the past decade, accumulating evidence has underscored the importance of the tumour microenvironment (TME), including tumour cells, stromal cells, immune cells, and various secreted factors, which collectively influence tumour growth, invasion, and responses to therapeutic agents. Immune cells within the TME are now widely acknowledged to play pivotal roles in tumour development and treatment. While some perspectives have posited that immune cells within the TME facilitate tumour progression and confer resistance to therapeutic interventions, contrasting conclusions also exist. Affirmative and negative conclusions appear to be context dependent, and a unified consensus has yet to be reached. The burgeoning body of research on the relationship between the gut microbiota and tumours in recent years has led to a growing understanding. Most studies have indicated that specific components of the gut microbiota, such as unique bacterial communities or specific secretory factors, play diverse roles in regulating immune cells within the TME, thereby influencing the prognosis and outcomes of cancer treatments. A detailed understanding of these factors could provide novel insights into the TME and cancer therapy. In this study, we aimed to synthesise information on the interactions between the gut microbiota and immune cells within the TME, providing an in-depth exploration of the potential guiding implications for future cancer therapies.
Collapse
Affiliation(s)
- Chunxiao Liu
- Department of Gastroenterological Surgery, Hengqin Hospital, First Affiliated Hospital of Guangzhou Medical University, No. 118 Baoxing Road, Hengqin, Guangdong, 519031, China
| | - Lingfeng Fu
- Department of Gastroenterological Surgery, Hengqin Hospital, First Affiliated Hospital of Guangzhou Medical University, No. 118 Baoxing Road, Hengqin, Guangdong, 519031, China
| | - Yuxin Wang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, No. 1838, North Guangzhou Avenue, Guangzhou, Guangdong, 510515, China.
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Weijun Yang
- Department of Gastroenterological Surgery, Hengqin Hospital, First Affiliated Hospital of Guangzhou Medical University, No. 118 Baoxing Road, Hengqin, Guangdong, 519031, China.
| |
Collapse
|
114
|
Munteanu C, Mârza SM, Papuc I. The immunomodulatory effects of vitamins in cancer. Front Immunol 2024; 15:1464329. [PMID: 39434876 PMCID: PMC11491384 DOI: 10.3389/fimmu.2024.1464329] [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: 07/13/2024] [Accepted: 09/17/2024] [Indexed: 10/23/2024] Open
Abstract
Nutrition may affect animal health due to the strong link between them. Also, diets improve the healing process in various disease states. Cancer is a disease, where the harmful consequences of tumors severely impair the body. The information regarding the evolution of this disease is extrapolated from human to animal because there are few specific studies regarding nutritional needs in animals with cancer. Thus, this paper aims to review the literature regarding the immunomodulatory effects of vitamins in mammal cancer. An adequate understanding of the metabolism and requirements of nutrients for mammals is essential to ensuring their optimal growth, development, and health, regardless of their food sources. According to these: 1) Some species are highly dependent on vitamin D from food, so special attention must be paid to this aspect. Calcitriol/VDR signaling can activate pro-apoptotic proteins and suppress anti-apoptotic ones. 2) Nitric oxide (NO) production is modulated by vitamin E through inhibiting transcription nuclear factor kappa B (NF-κB) activation. 3) Thiamine supplementation could be responsible for the stimulation of tumor cell proliferation, survival, and resistance to chemotherapy. 4) Also, it was found that the treatment with NO-Cbl in dogs is a viable anti-cancer therapy that capitalizes on the tumor-specific properties of the vitamin B12 receptor. Therefore, diets should contain the appropriate class of compounds in adequate proportions. Also, the limitations of this paper are that some vitamins are intensively studied and at the same time regarding others, there is a lack of information, especially in animals. Therefore, some subsections are longer and more heavily debated than others.
Collapse
Affiliation(s)
- Camelia Munteanu
- Department of Plant Culture, Faculty of Agriculture, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, ;Romania
| | - Sorin Marian Mârza
- Faculty of Veterinary Medicine, University of Agricultural Science and Veterinary Medicine, Cluj-Napoca, ;Romania
| | - Ionel Papuc
- Faculty of Veterinary Medicine, University of Agricultural Science and Veterinary Medicine, Cluj-Napoca, ;Romania
| |
Collapse
|
115
|
Wang L, Zhang L, Zhang Z, Wu P, Zhang Y, Chen X. Advances in targeting tumor microenvironment for immunotherapy. Front Immunol 2024; 15:1472772. [PMID: 39421736 PMCID: PMC11484021 DOI: 10.3389/fimmu.2024.1472772] [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: 07/30/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
The tumor microenvironment (TME) provides essential conditions for the occurrence, invasion, and spread of cancer cells. Initial research has uncovered immunosuppressive properties of the TME, which include low oxygen levels (hypoxia), acidic conditions (low pH), increased interstitial pressure, heightened permeability of tumor vasculature, and an inflammatory microenvironment. The presence of various immunosuppressive components leads to immune evasion and affects immunotherapy efficacy. This indicates the potential value of targeting the TME in cancer immunotherapy. Therefore, TME remodeling has become an effective method for enhancing host immune responses against tumors. In this study, we elaborate on the characteristics and composition of the TME and how it weakens immune surveillance and summarize targeted therapeutic strategies for regulating the TME.
Collapse
Affiliation(s)
- Lugang Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liubo Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhen Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Wu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Engineering Key Laboratory for Cell Therapy of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xinfeng Chen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
116
|
Haddad A, Holder AM. Microbiome and Immunotherapy for Melanoma: Are We Ready for Clinical Application? Hematol Oncol Clin North Am 2024; 38:1061-1070. [PMID: 38908958 DOI: 10.1016/j.hoc.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
The microbiome plays a substantial role in the efficacy of immune checkpoint blockade (ICB) in patients with metastatic melanoma. While the exact gut microbiome composition and the pathways involved in this interaction are not clearly delineated, novel studies and ongoing clinical trials are likely to reveal findings applicable to the clinical setting for the prediction and optimization of response to ICB. Nevertheless, lifestyle modifications, including high fiber diet, avoidance of unnecessary antibiotic prescriptions, and careful use of probiotics may be helpful to optimize the "health" of the gut microbiome and potentially enhance response to ICB in patients with melanoma.
Collapse
Affiliation(s)
- Antony Haddad
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1484, Houston, TX 77030, USA. https://twitter.com/Haddad_Antony
| | - Ashley M Holder
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1484, Houston, TX 77030, USA.
| |
Collapse
|
117
|
Fleming N. Your diet can change your immune system - here's how. Nature 2024; 634:528-531. [PMID: 39415064 DOI: 10.1038/d41586-024-03334-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
|
118
|
Belaid A, Roméo B, Rignol G, Benzaquen J, Audoin T, Vouret-Craviari V, Brest P, Varraso R, von Bergen M, Hugo Marquette C, Leroy S, Mograbi B, Hofman P. Impact of the Lung Microbiota on Development and Progression of Lung Cancer. Cancers (Basel) 2024; 16:3342. [PMID: 39409962 PMCID: PMC11605235 DOI: 10.3390/cancers16193342] [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: 08/30/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 12/01/2024] Open
Abstract
The past several years have provided a more profound understanding of the role of microbial species in the lung. The respiratory tract is a delicate ecosystem of bacteria, fungi, parasites, and viruses. Detecting microbial DNA, pathogen-associated molecular patterns (PAMPs), and metabolites in sputum is poised to revolutionize the early diagnosis of lung cancer. The longitudinal monitoring of the lung microbiome holds the potential to predict treatment response and side effects, enabling more personalized and effective treatment options. However, most studies into the lung microbiota have been observational and have not adequately considered the impact of dietary intake and air pollutants. This gap makes it challenging to establish a direct causal relationship between environmental exposure, changes in the composition of the microbiota, lung carcinogenesis, and tumor progression. A holistic understanding of the lung microbiota that considers both diet and air pollutants may pave the way to improved prevention and management strategies for lung cancer.
Collapse
Affiliation(s)
- Amine Belaid
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
| | - Barnabé Roméo
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
| | - Guylène Rignol
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
- Laboratory of Clinical and Experimental Pathology (LPCE), Biobank (BB-0033-00025), Centre Hospitalier Universitaire (CHU) de Nice, FHU OncoAge, IHU RespirERA, 06000 Nice, France
| | - Jonathan Benzaquen
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
- Centre Hospitalier Universitaire (CHU) de Nice, Department of Pulmonary Medicine and Thoracic Oncology, FHU OncoAge, IHU RespirERA, 06000 Nice, France
| | - Tanguy Audoin
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
| | - Valérie Vouret-Craviari
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
| | - Patrick Brest
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
| | - Raphaëlle Varraso
- Université Paris-Saclay, Équipe d’Épidémiologie Respiratoire Intégrative, CESP, INSERM, 94800 Villejuif, France;
| | - Martin von Bergen
- Helmholtz Centre for Environmental Research GmbH—UFZ, Department of Molecular Systems Biology, Institute of Biochemistry, Faculty of Life Sciences, University of Leipzig, 04109 Leipzig, Germany;
| | - Charles Hugo Marquette
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
- Centre Hospitalier Universitaire (CHU) de Nice, Department of Pulmonary Medicine and Thoracic Oncology, FHU OncoAge, IHU RespirERA, 06000 Nice, France
| | - Sylvie Leroy
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
- Centre Hospitalier Universitaire (CHU) de Nice, Department of Pulmonary Medicine and Thoracic Oncology, FHU OncoAge, IHU RespirERA, 06000 Nice, France
| | - Baharia Mograbi
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
| | - Paul Hofman
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
- Laboratory of Clinical and Experimental Pathology (LPCE), Biobank (BB-0033-00025), Centre Hospitalier Universitaire (CHU) de Nice, FHU OncoAge, IHU RespirERA, 06000 Nice, France
| |
Collapse
|
119
|
Missiego-Beltrán J, Beltrán-Velasco AI. The Role of Microbial Metabolites in the Progression of Neurodegenerative Diseases-Therapeutic Approaches: A Comprehensive Review. Int J Mol Sci 2024; 25:10041. [PMID: 39337526 PMCID: PMC11431950 DOI: 10.3390/ijms251810041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
The objective of this review is to provide a comprehensive examination of the role of microbial metabolites in the progression of neurodegenerative diseases, as well as to investigate potential therapeutic interventions targeting the microbiota. A comprehensive literature search was conducted across the following databases: PubMed, Scopus, Web of Science, ScienceDirect, and Wiley. Key terms related to the gut microbiota, microbial metabolites, neurodegenerative diseases, and specific metabolic products were used. The review included both preclinical and clinical research articles published between 2000 and 2024. Short-chain fatty acids have been demonstrated to play a crucial role in modulating neuroinflammation, preserving the integrity of the blood-brain barrier, and influencing neuronal plasticity and protection. Furthermore, amino acids and their derivatives have been demonstrated to exert a significant influence on CNS function. These microbial metabolites impact CNS health by regulating intestinal permeability, modulating immune responses, and directly influencing neuroinflammation and oxidative stress, which are integral to neurodegenerative diseases. Therapeutic strategies, including prebiotics, probiotics, dietary modifications, and fecal microbiota transplantation have confirmed the potential to restore microbial balance and enhance the production of neuroprotective metabolites. Furthermore, novel drug developments based on microbial metabolites present promising therapeutic avenues. The gut microbiota and its metabolites represent a promising field of research with the potential to advance our understanding of and develop treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Ana Isabel Beltrán-Velasco
- NBC Group, Psychology Department, School of Life and Nature Sciences, Nebrija University, 28015 Madrid, Spain;
| |
Collapse
|
120
|
Messaoudene M, Ferreira S, Saint-Lu N, Ponce M, Truntzer C, Boidot R, Le Bescop C, Loppinet T, Corbel T, Féger C, Bertrand K, Elkrief A, Isaksen M, Vitry F, Sablier-Gallis F, Andremont A, Bod L, Ghiringhelli F, de Gunzburg J, Routy B. The DAV132 colon-targeted adsorbent does not interfere with plasma concentrations of antibiotics but prevents antibiotic-related dysbiosis: a randomized phase I trial in healthy volunteers. Nat Commun 2024; 15:8083. [PMID: 39278946 PMCID: PMC11402973 DOI: 10.1038/s41467-024-52373-8] [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: 06/05/2024] [Accepted: 09/05/2024] [Indexed: 09/18/2024] Open
Abstract
The deleterious impact of antibiotics (ATB) on the microbiome negatively influences immune checkpoint inhibitors (ICI) response in patients with cancer. We conducted a randomized phase I study (EudraCT:2019-A00240-57) with 148 healthy volunteers (HV) to test two doses of DAV132, a colon-targeted adsorbent, alongside intravenous ceftazidime-avibactam (CZA), piperacillin-tazobactam (PTZ) or ceftriaxone (CRO) and a group without ATB. The primary objective of the study was to assess the effect of DAV132 on ATB plasma concentrations and both doses of DAV132 did not alter ATB levels. Secondary objectives included safety, darkening of the feces, and fecal ATB concentrations. DAV132 was well tolerated, with no severe toxicity and similar darkening at both DAV132 doses. DAV132 led to significant decrease in CZA or PTZ feces concentration. When co-administered with CZA or PTZ, DAV132 preserved microbiome diversity, accelerated recovery to baseline composition and protected key commensals. Fecal microbiota transplantation (FMT) in preclinical cancer models in female mice from HV treated with CZA or PTZ alone inhibited anti-PD-1 response, while transplanted samples from HV treated with ATB + DAV132 circumvented resistance to anti-PD-1. This effect was linked to activated CD8+ T cell populations in the tumor microenvironment. DAV132 represents a promising strategy for overcoming ATB-related dysbiosis and further studies are warranted to evaluate its efficacy in cancer patients.
Collapse
Affiliation(s)
- Meriem Messaoudene
- Axe Cancer, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | | | | | - Mayra Ponce
- Axe Cancer, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Caroline Truntzer
- Platform of Transfer in Biological Oncology, Georges François Leclerc Cancer Center-Unicancer, Dijon, France
- UMR INSERM 1231, Dijon, France
| | - Romain Boidot
- Molecular Biology, Georges François Leclerc Cancer Center-Unicancer, Dijon, France
| | | | | | | | - Céline Féger
- Da Volterra, Paris, France
- Medical, EMI Biotech, Paris, France
| | | | - Arielle Elkrief
- Axe Cancer, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Hemato-Oncology Division, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | | | | | | | | | - Lloyd Bod
- Krantz Family Cancer Center, Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - François Ghiringhelli
- Department of Medical Oncology, Georges François Leclerc Cancer Center-Unicancer, Dijon, France
| | | | - Bertrand Routy
- Axe Cancer, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.
- Hemato-Oncology Division, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada.
| |
Collapse
|
121
|
Du Y, Wang Q, Zheng Z, Zhou H, Han Y, Qi A, Jiao L, Gong Y. Gut microbiota influence on lung cancer risk through blood metabolite mediation: from a comprehensive Mendelian randomization analysis and genetic analysis. Front Nutr 2024; 11:1425802. [PMID: 39323566 PMCID: PMC11423778 DOI: 10.3389/fnut.2024.1425802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Background Gut microbiota (GM) and metabolic alterations play pivotal roles in lung cancer (LC) development and host genetic variations are known to contribute to LC susceptibility by modulating the GM. However, the causal links among GM, metabolite, host genes, and LC remain to be fully delineated. Method Through bidirectional MR analyses, we examined the causal links between GM and LC, and utilized two-step mediation analysis to identify potential mediating blood metabolite. We employed diverse MR methods, including inverse-variance-weighted (IVW), weighted median, MR-Egger, weighted mode, and simple mode, to ensure a robust examination of the data. MR-Egger intercept test, Radial MR, MR-PRESSO, Cochran Q test and Leave-one-out (LOO) analysis were used for sensitivity analyses. Analyses were adjusted for smoking, alcohol intake frequency and air pollution. Linkage disequilibrium score regression and Steiger test were used to probe genetic causality. The study also explored the association between specific host genes and the abundance of gut microbes in LC patients. Results The presence of Bacteroides clarus was associated with an increased risk of LC (odds ratio [OR] = 1.07, 95% confidence interval [CI]: 1.03-1.11, p = 0.012), whereas the Eubacteriaceae showed a protective effect (OR = 0.82, 95% CI: 0.75-0.89, p = 0.001). These findings remained robust after False Discovery Rate (FDR) correction. Our mediator screening identified 13 blood metabolites that significantly influence LC risk after FDR correction, underscoring cystine and propionylcarnitine in reducing LC risk, while linking specific lipids and hydroxy acids to an increased risk. Our two-step mediation analysis demonstrated that the association between the bacterial pathway of synthesis of guanosine ribonucleotides and LC was mediated by Fructosyllysine, with mediated proportions of 11.38% (p = 0.037). LDSC analysis confirmed the robustness of these associations. Our study unveiled significant host genes ROBO2 may influence the abundance of pathogenic gut microbes in LC patients. Metabolic pathway analysis revealed glutathione metabolism and glutamate metabolism are the pathways most enriched with significant metabolites related to LC. Conclusion These findings underscore the importance of GM in the development of LC, with metabolites partly mediating this effect, and provide dietary and lifestyle recommendations for high-risk lung cancer populations.
Collapse
Affiliation(s)
- Yizhao Du
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qin Wang
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zongmei Zheng
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hailun Zhou
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yang Han
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ao Qi
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lijing Jiao
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Translational Cancer Research for Integrated Chinese and Western Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yabin Gong
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
122
|
Zitvogel L, Fidelle M, Kroemer G. Long-distance microbial mechanisms impacting cancer immunosurveillance. Immunity 2024; 57:2013-2029. [PMID: 39151425 DOI: 10.1016/j.immuni.2024.07.020] [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: 05/30/2024] [Revised: 07/13/2024] [Accepted: 07/21/2024] [Indexed: 08/19/2024]
Abstract
The intestinal microbiota determines immune responses against extraintestinal antigens, including tumor-associated antigens. Indeed, depletion or gross perturbation of the microbiota undermines the efficacy of cancer immunotherapy, thereby compromising the clinical outcome of cancer patients. In this review, we discuss the long-distance effects of the gut microbiota and the mechanisms governing antitumor immunity, such as the translocation of intestinal microbes into tumors, migration of leukocyte populations from the gut to the rest of the body, including tumors, as well as immunomodulatory microbial products and metabolites. The relationship between these pathways is incompletely understood, in particular the significance of the tumor microbiota with respect to the identification of host and/or microbial products that regulate the egress of bacteria and immunocytes toward tumor beds.
Collapse
Affiliation(s)
- Laurence Zitvogel
- Gustave Roussy Cancer Campus, Villejuif, France; Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR 1015, ClinicObiome, Équipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; Université Paris-Saclay, Ile-de-France, France; Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS), Villejuif, France.
| | - Marine Fidelle
- Gustave Roussy Cancer Campus, Villejuif, France; Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR 1015, ClinicObiome, Équipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; Université Paris-Saclay, Ile-de-France, France
| | - Guido Kroemer
- Gustave Roussy Cancer Campus, Villejuif, France; Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France; Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
123
|
Le Ngoc K, Pham TTH, Nguyen TK, Huong PT. Pharmacomicrobiomics in precision cancer therapy: bench to bedside. Front Immunol 2024; 15:1428420. [PMID: 39315107 PMCID: PMC11416994 DOI: 10.3389/fimmu.2024.1428420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
The burgeoning field of pharmacomicrobiomics offers promising insights into the intricate interplay between the microbiome and cancer, shaping responses to diverse treatment modalities. This review aims to analyze the molecular mechanisms underlying interactions between distinct microbiota types and cancer, as well as their influence on treatment outcomes. We explore how the microbiome impacts antitumor immunity, and response to chemotherapy, immunotherapy, and radiation therapy, unveiling its multifaceted roles in cancer progression and therapy resistance. Moreover, we discuss the challenges hindering the development of microbiome-based interventions in cancer therapy, including standardization, validation, and clinical translation. By synthesizing clinical evidence, we underscore the transformative potential of harnessing pharmacomicrobiomics in guiding cancer treatment decisions, paving the way for improved patient outcomes in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Phung Thanh Huong
- Faculty of Biotechnology, Hanoi University of Pharmacy,
Hanoi, Vietnam
| |
Collapse
|
124
|
Xu Y, He C, Xi Y, Zhang Y, Bai Y. Gut microbiota and immunosenescence in cancer. Semin Cancer Biol 2024; 104-105:32-45. [PMID: 39127266 DOI: 10.1016/j.semcancer.2024.07.004] [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: 06/25/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024]
Abstract
Cancer is generally defined as a disease of aging. With aging, the composition, diversity and functional characteristics of the gut microbiota occur changes, with a decline of beneficial commensal microbes triggered by intrinsic and extrinsic factors (e.g., diet, drugs and chronic health conditions). Nowadays, dysbiosis of the gut microbiota is recognized as a hallmark of cancer. At the same time, aging is accompanied by changes in innate and adaptive immunity, known as immunosenescence, as well as chronic low-grade inflammation, known as inflammaging. The elevated cancer incidence and mortality in the elderly are linked with aging-associated alterations in the gut microbiota that elicit systemic metabolic alterations, leading to immune dysregulation with potentially tumorigenic effects. The gut microbiota and immunosenescence might both affect the response to treatment in cancer patients. In-depth understanding of age-associated alterations in the gut microbiota and immunity will shed light on the risk of cancer development and progression in the elderly. Here, we describe the aging-associated changes of the gut microbiota in cancer, and review the evolving understanding of the gut microbiota-targeted intervention strategies. Furthermore, we summarize the knowledge on the cellular and molecular mechanisms of immunosenescence and its impact on cancer. Finally, we discuss the latest knowledge about the relationships between gut microbiota and immunosenescence, with implications for cancer therapy. Intervention strategies targeting the gut microbiota may attenuate inflammaging and rejuvenate immune function to provide antitumor benefits in elderly patients.
Collapse
Affiliation(s)
- Yaozheng Xu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110136, China; Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning 110136, China.
| | - Chuan He
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Ying Xi
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110136, China; Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning 110136, China.
| | - Yue Zhang
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110136, China; Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning 110136, China.
| | - Yibo Bai
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110136, China; Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning 110136, China.
| |
Collapse
|
125
|
Wang X, Gao X, Chen Y, Wu X, Shang Y, Zhang Z, Zhou S, Zhang H. Comparative analysis of the gut microbiome of ungulate species from Qinghai-Xizang plateau. Ecol Evol 2024; 14:e70251. [PMID: 39257880 PMCID: PMC11387016 DOI: 10.1002/ece3.70251] [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: 01/30/2024] [Revised: 08/09/2024] [Accepted: 08/16/2024] [Indexed: 09/12/2024] Open
Abstract
Several studies have investigated the gut bacterial composition of wild ungulates in the Qinghai-Xizang Plateau. However, the relationship between their gut microbiome dendrograms and their phylogenetic tree remains unclear. In this study, we analyzed 45 amplicons (V3-V4 region of the 16S rRNA gene) from five wild ungulates-Pseudois nayaur, Pantholops hodgsonii, Gazella subgutturosa, Bos grunniens, and Equus kiang-from the Qinghai-Xizang Plateau to clarify the relationship between their phylogenies and gut microbiome dendrograms. The unweighted pair group method with arithmetic mean analysis and hierarchical clustering analysis indicated that G. subgutturosa is closely related to P. nayaur; however, these results were inconsistent with their phylogenetic trees. Additionally, the indicator genera in the microbiome of each wild ungulate showed strong associations with the diets and habitats of their host. Thus, diet and space niche differentiation may primarily account for the differences between the gut microbiome characteristics of these wild ungulates and their phylogeny. In summary, our research provides insights into the evolutionary factors influencing the gut microbiome of wild ungulates in the Qinghai-Xizang Plateau.
Collapse
Affiliation(s)
- Xibao Wang
- College of Life SciencesQufu Normal UniversityQufuShandongChina
| | - Xiaodong Gao
- College of Life SciencesQufu Normal UniversityQufuShandongChina
| | - Yao Chen
- College of Life SciencesQufu Normal UniversityQufuShandongChina
| | - Xiaoyang Wu
- College of Life SciencesQufu Normal UniversityQufuShandongChina
| | - Yongquan Shang
- College of Life SciencesQufu Normal UniversityQufuShandongChina
| | - Zhihao Zhang
- College of Life SciencesQufu Normal UniversityQufuShandongChina
| | - Shengyang Zhou
- College of Life SciencesQufu Normal UniversityQufuShandongChina
| | - Honghai Zhang
- College of Life SciencesQufu Normal UniversityQufuShandongChina
| |
Collapse
|
126
|
Tamer F, Kekilli M. Exploring the therapeutic potential of topical probiotics in dermatological diseases: a comprehensive review of clinical studies. J Dtsch Dermatol Ges 2024; 22:1195-1204. [PMID: 39164814 DOI: 10.1111/ddg.15465] [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: 12/05/2023] [Accepted: 04/23/2024] [Indexed: 08/22/2024]
Abstract
The gastrointestinal tract may contribute to the regulation of systemic inflammation and skin diseases due to the balance between the pathogenic, opportunistic, and beneficial bacterial species it contains. External supplementation of beneficial bacteria, besides its known positive effect in the treatment of digestive system diseases, also has different favorable effects such as accelerated wound healing, suppression of inflammation, lower infection risk, and reduced antibiotic requirement. It has been reported that oral use of probiotics may be effective in the treatment of skin diseases such as acne, psoriasis, and atopic dermatitis. Furthermore, topical administration of probiotics may create a positive bacterial balance, eliminate pathological conditions, and thus have a favorable impact on the management of skin diseases. Interest in the effect of the skin microbiome and topical probiotics in the treatment of skin diseases has increased recently. Herein, 21 clinical studies and a case report in the PubMed database on the use of topical probiotics in dermatological diseases were evaluated.
Collapse
Affiliation(s)
- Funda Tamer
- Department of Dermatology, Gazi University School of Medicine, Ankara, Turkey
| | - Murat Kekilli
- Department of Gastroenterology, Gazi University School of Medicine, Department of Gastroenterology, Ankara, Turkey
| |
Collapse
|
127
|
Ebrahimi H, Dizman N, Meza L, Malhotra J, Li X, Dorff T, Frankel P, Llamas-Quitiquit M, Hsu J, Zengin ZB, Alcantara M, Castro D, Mercier B, Chawla N, Chehrazi-Raffle A, Barragan-Carrillo R, Jaime-Casas S, Govindarajan A, Gillece J, Trent J, Lee PP, Parks TP, Takahashi M, Hayashi A, Kortylewski M, Caporaso JG, Lee K, Tripathi A, Pal SK. Cabozantinib and nivolumab with or without live bacterial supplementation in metastatic renal cell carcinoma: a randomized phase 1 trial. Nat Med 2024; 30:2576-2585. [PMID: 38942995 PMCID: PMC11405272 DOI: 10.1038/s41591-024-03086-4] [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: 02/04/2024] [Accepted: 05/23/2024] [Indexed: 06/30/2024]
Abstract
Supplementation with CBM588, a bifidogenic live bacterial product, has been associated with improved clinical outcomes in persons with metastatic renal cell carcinoma (mRCC) receiving nivolumab and ipilimumab. However, its effect on those receiving tyrosine kinase inhibitor-based combinations is unknown. In this open-label, randomized, investigator-initiated, phase 1 study, 30 participants with locally advanced or mRCC with histological confirmation of clear cell, papillary or sarcomatoid component were randomized in a 2:1 fashion to receive cabozantinib (an inhibitor of vascular endothelial growth factor receptor, MET and AXL) and nivolumab (anti-programmed cell death protein 1) with or without CBM588 as first-line treatment. Metagenomic sequencing was performed on stool samples to characterize their gut microbiome at baseline and 13 weeks into treatment. The primary endpoint was a change in the relative abundance of Bifidobacterium spp.; secondary endpoints included objective response rate (ORR), progression-free survival (PFS) and toxicity profile. The primary endpoint of the study was not met and the addition of CBM588 to cabozantinib and nivolumab did not result in a difference in the relative abundance of Bifidobacterium spp. or alpha diversity (as measured by the Shannon index). However, ORR was significantly higher in participants treated with CBM588 compared to those in the control arm (14 of 19, 74% versus 2 of 10, 20%; P = 0.01). PFS at 6 months was 84% (16 of 19) and 60% (6 of 10) in the experimental and control arms, respectively. No significant difference in toxicity profile was seen between the study arms. Our results provide a preliminary signal of improved clinical activity with CBM588 in treatment-naive participants with mRCC receiving cabozantinib and nivolumab. Further investigation is needed to confirm these findings and better characterize the underlying mechanism driving this effect.ClinicalTrials.gov identifier: NCT05122546.
Collapse
Affiliation(s)
- Hedyeh Ebrahimi
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Nazli Dizman
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
- MD Anderson Cancer Center, Houston, TX, USA
| | - Luis Meza
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
- Yale University School of Medicine, New Haven, CT, USA
| | - Jasnoor Malhotra
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Xiaochen Li
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Tanya Dorff
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Paul Frankel
- Department of Biostatistics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | - Joann Hsu
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Zeynep B Zengin
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
- Yale University School of Medicine, New Haven, CT, USA
| | - Marice Alcantara
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Daniela Castro
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Benjamin Mercier
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Neal Chawla
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Alex Chehrazi-Raffle
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | - Salvador Jaime-Casas
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Ameish Govindarajan
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - John Gillece
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Jeffrey Trent
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Peter P Lee
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | | | | | - Marcin Kortylewski
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | - Keehoon Lee
- Translational Genomics Research Institute (TGen), Flagstaff, AZ, USA
| | - Abhishek Tripathi
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| | - Sumanta K Pal
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| |
Collapse
|
128
|
Verhaert MAM, Aspeslagh S. Immunotherapy efficacy and toxicity: Reviewing the evidence behind patient implementable strategies. Eur J Cancer 2024; 209:114235. [PMID: 39059186 DOI: 10.1016/j.ejca.2024.114235] [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: 04/11/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Abstract
The use of immune checkpoint inhibitors (ICI) in cancer treatment is expanding, offering promising outcomes but with an important risk of immune-related adverse events (irAEs). These events, stemming from an overstimulated immune system attacking healthy cells, can necessitate immunosuppressant treatment, disrupt treatment courses, and impact patients' quality of life. The analysis of ICI efficacy data has led to a better understanding of the characteristics of responders. Similarly, we are gaining clearer insights into the characteristics of patients who develop irAEs, prompting an increasing emphasis on modifiable factors associated with irAE risk. These factors include lifestyle choices and the composition of the gut microbiome. Despite comprehensive reviews exploring the microbiome's role in therapy efficacy, understanding its connection with immune-related toxicity remains incomplete. While endeavours to identify predictive biomarkers continue, lifestyle modifications emerge as a promising avenue for enhancing treatment outcomes. This review consolidates the current evidence regarding the impact of the gut microbiome on irAE occurrence. Furthermore, it focuses on actionable strategies for mitigating these adverse events, elucidating the evidence supporting dietary adjustments, supplementation, medication management, and physical activity. With the expanding range of indications for ICI therapy, a significant proportion of oncology patients, including those in early disease stages, are now exposed to these treatments. Acknowledging the importance of averting irAEs in this context, our review offers timely insights crucial for addressing the evolving challenges associated with immunotherapy across diverse oncological settings.
Collapse
Affiliation(s)
- Marthe August Marianne Verhaert
- Department of Medical Oncology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium.
| | - Sandrine Aspeslagh
- Department of Medical Oncology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium; Department of Internal Medicine, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
| |
Collapse
|
129
|
Gerasimova Y, Ali H, Nadeem U. Challenges for pathologists in implementing clinical microbiome diagnostic testing. J Pathol Clin Res 2024; 10:e70002. [PMID: 39289163 PMCID: PMC11407905 DOI: 10.1002/2056-4538.70002] [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: 12/21/2023] [Revised: 08/11/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024]
Abstract
Recent research has established that the microbiome plays potential roles in the pathogenesis of numerous chronic diseases, including carcinomas. This discovery has led to significant interest in clinical microbiome testing among physicians, translational investigators, and the lay public. As novel, inexpensive methodologies to interrogate the microbiota become available, research labs and commercial vendors have offered microbial assays. However, these tests still have not infiltrated the clinical laboratory space. Here, we provide an overview of the challenges of implementing microbiome testing in clinical pathology. We discuss challenges associated with preanalytical and analytic sample handling and collection that can influence results, choosing the appropriate testing methodology for the clinical context, establishing reference ranges, interpreting the data generated by testing and its value in making patient care decisions, regulation, and cost considerations of testing. Additionally, we suggest potential solutions for these problems to expedite the establishment of microbiome testing in the clinical laboratory.
Collapse
Affiliation(s)
- Yulia Gerasimova
- Department of Infectious Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Haroon Ali
- Department of Medicine, Woodland Heights Medical Center, Lufkin, TX, USA
| | - Urooba Nadeem
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
130
|
Ryan T, Ling S, Trinh A, Segal JP. The role of the microbiome in immune checkpoint inhibitor colitis and hepatitis. Best Pract Res Clin Gastroenterol 2024; 72:101945. [PMID: 39645281 DOI: 10.1016/j.bpg.2024.101945] [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: 05/20/2024] [Accepted: 08/05/2024] [Indexed: 12/09/2024]
Abstract
Immune checkpoint inhibitors have revolutionised management for a variety of different types of malignancies. However, gastrointestinal adverse effects, in particular colitis and hepatitis, are relatively common with up to 30 % of patients being affected. The gut microbiome has emerged as a potential contributor to both the effectiveness of immune checkpoint inhibitors and their side effects. This review will attempt to examine the impact the microbiome has on adverse effects as a result of immune checkpoint inhibitors as well as the potential for manipulation of the microbiome as a form of management for immune mediated colitis.
Collapse
Affiliation(s)
- Thomas Ryan
- Faculty of Medicine, University of Melbourne, Melbourne, Australia.
| | - Sophia Ling
- Department of Gastroenterology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Andrew Trinh
- Department of Gastroenterology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Jonathan P Segal
- Faculty of Medicine, University of Melbourne, Melbourne, Australia; Department of Gastroenterology, Peter MacCallum Cancer Centre, Melbourne, Australia
| |
Collapse
|
131
|
Xia L, Zhu X, Wang Y, Lu S. The gut microbiota improves the efficacy of immune-checkpoint inhibitor immunotherapy against tumors: From association to cause and effect. Cancer Lett 2024; 598:217123. [PMID: 39033797 DOI: 10.1016/j.canlet.2024.217123] [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: 05/08/2024] [Revised: 06/20/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Immune-checkpoint inhibitors (ICIs), including anti-PD-1/PD-L1 therapeutic antibodies, have markedly enhanced survival across numerous cancer types. However, the limited number of patients with durable benefits creates an urgent need to identify response biomarkers and to develop novel strategies so as to improve response. It is widely recognized that the gut microbiome is a key mediator in shaping immunity. Additionally, the gut microbiome shows significant potential in predicting the response to and enhancing the efficacy of ICI immunotherapy against cancer. Recent studies encompassing mechanistic analyses and clinical trials of microbiome-based therapy have shown a cause-and-effect relationship between the gut microbiome and the modulation of the ICI immunotherapeutic response, greatly contributing to the establishment of novel strategies that will improve response and overcome resistance to ICI treatment. In this review, we outline the current state of research advances and discuss the future directions of utilizing the gut microbiome to enhance the efficacy of ICI immunotherapy against tumors.
Collapse
Affiliation(s)
- Liliang Xia
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, PR China
| | - Xiaokuan Zhu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, PR China
| | - Ying Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, PR China.
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, PR China.
| |
Collapse
|
132
|
Li X, Shang S, Wu M, Song Q, Chen D. Gut microbial metabolites in lung cancer development and immunotherapy: Novel insights into gut-lung axis. Cancer Lett 2024; 598:217096. [PMID: 38969161 DOI: 10.1016/j.canlet.2024.217096] [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: 04/03/2024] [Revised: 06/11/2024] [Accepted: 06/28/2024] [Indexed: 07/07/2024]
Abstract
Metabolic derivatives of numerous microorganisms inhabiting the human gut can participate in regulating physiological activities and immune status of the lungs through the gut-lung axis. The current well-established microbial metabolites include short-chain fatty acids (SCFAs), tryptophan and its derivatives, polyamines (PAs), secondary bile acids (SBAs), etc. As the study continues to deepen, the critical function of microbial metabolites in the occurrence and treatment of lung cancer has gradually been revealed. Microbial derivates can enter the circulation system to modulate the immune microenvironment of lung cancer. Mechanistically, oncometabolites damage host DNA and promote the occurrence of lung cancer, while tumor-suppresive metabolites directly affect the immune system to combat the malignant properties of cancer cells and even show considerable application potential in improving the efficacy of lung cancer immunotherapy. Considering the crosstalk along the gut-lung axis, in-depth exploration of microbial metabolites in patients' feces or serum will provide novel guidance for lung cancer diagnosis and treatment selection strategies. In addition, targeted therapeutics on microbial metabolites are expected to overcome the bottleneck of lung cancer immunotherapy and alleviate adverse reactions, including fecal microbiota transplantation, microecological preparations, metabolite synthesis and drugs targeting metabolic pathways. In summary, this review provides novel insights and explanations on the intricate interplay between gut microbial metabolites and lung cancer development, and immunotherapy through the lens of the gut-lung axis, which further confirms the possible translational potential of the microbiome metabolome in lung cancer treatment.
Collapse
Affiliation(s)
- Xinpei Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shijie Shang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Wu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Qian Song
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| | - Dawei Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
133
|
Zhang Y, Zhou C, Tian B, Xu J, Wang X, Dai H, Wang H, Xu F, Wang C. 3D printed spiral tube-like cellulose scaffold for oral delivery of probiotics. SCIENCE ADVANCES 2024; 10:eadp3654. [PMID: 39178258 PMCID: PMC11343031 DOI: 10.1126/sciadv.adp3654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/19/2024] [Indexed: 08/25/2024]
Abstract
Introducing specific strains of probiotics into the gut microbiome is a promising way to modulate the intestinal microbiome to treat various health conditions clinically. However, oral probiotics typically have a temporary or limited impact on the gut microbiome and overall health benefits. Here, we reported a 3D printed cellulose-derived spiral tube-like scaffold that enabled high efficacy of the oral delivery of probiotics. Benefiting from the unique surface pattern, this system can effectively extend the retention time of loaded probiotics in the gut without invading nearby tissues, provide a favorable environment for the survival and long-term colonization of loaded probiotics, and influence the intestinal ecosystem as a dietary fiber after degradation. We demonstrate Roseburia intestinalis-loaded scaffold exerts noticeable impacts on the regulation of the gut microbiome to treat various gut-related diseases, including obesity and inflammatory bowel disease; thus, we provide a universal platform for oral delivery of probiotics.
Collapse
Affiliation(s)
- Yue Zhang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Chenmeng Zhou
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Bo Tian
- The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, China
| | - Jialu Xu
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Xiaoyu Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Huaxing Dai
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Heng Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Fang Xu
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Chao Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
134
|
Pérez-Peláez B, Jiménez-Cortegana C, de la Cruz-Merino L, Sánchez-Margalet V. Role of Nutrients Regulating Myeloid Derived Suppressor Cells in Cancer: A Scoping Review. Curr Issues Mol Biol 2024; 46:9286-9297. [PMID: 39329901 PMCID: PMC11429620 DOI: 10.3390/cimb46090549] [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: 08/02/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/28/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are immature cells with an immunosuppressive function. MDSCs have been related to inflammation in many settings, including infections, transplantation, obesity, aging, or cancer. In oncological settings, MDSCs participate in tumor immunoescape, growth, and metastasis. Certain nutrients can modify chronic inflammation by their interaction with MDSCs. Therefore, the possible influence of certain nutrients on immune surveillance by their actions on MDSCs and how this may affect the prognosis of cancer patients were evaluated in this scoping review. We identified seven papers, six of which were murine model studies and only one was a human clinical trial. Globally, a significant reduction in cancer growth and progression was observed after achieving a reduction in both MDSCs and their immunosuppressive ability with nutrients such as selected vegetables, icaritin, retinoic acid, curdlan, active vitamin D, soy isoflavones, and green tea. In conclusion, the consumption of certain nutrients may have effects on MDSCs, with beneficial results not only in the prevention of tumor development and growth but also in improving patients' response.
Collapse
Affiliation(s)
- Beatriz Pérez-Peláez
- Department of Medical Biochemistry and Molecular Biology and Immunology, School of Medicine, Clinical Biochemistry Service, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain; (B.P.-P.); (C.J.-C.)
| | - Carlos Jiménez-Cortegana
- Department of Medical Biochemistry and Molecular Biology and Immunology, School of Medicine, Clinical Biochemistry Service, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain; (B.P.-P.); (C.J.-C.)
| | - Luis de la Cruz-Merino
- Department of Medicine, School of Medicine, Clinical Oncology Service, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain;
- Institute of Biomedicine of Seville, Virgen Macarena University Hospital, CSIC, University of Seville, 41013 Seville, Spain
| | - Víctor Sánchez-Margalet
- Department of Medicine, School of Medicine, Clinical Oncology Service, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain;
- Institute of Biomedicine of Seville, Virgen Macarena University Hospital, CSIC, University of Seville, 41013 Seville, Spain
| |
Collapse
|
135
|
Zhang M, Bzura A, Baitei EY, Zhou Z, Spicer JB, Poile C, Rogel J, Branson A, King A, Barber S, Kamata T, Dzialo J, Harber J, Greystoke A, Nusrat N, Faulkner D, Sun Q, Nolan L, Hahne JC, Scotland M, Walter H, Darlison L, Morgan B, Bajaj A, Brookes C, Hollox EJ, Lubawska D, Jama M, Griffiths G, Nakas A, Kutywayo K, Luo JL, Klampatsa A, Cooper A, Halder K, Wells-Jordan P, Zhou H, Dudbridge F, Thomas A, Richards CJ, Pritchard C, Yang H, Barer M, Fennell DA. A gut microbiota rheostat forecasts responsiveness to PD-L1 and VEGF blockade in mesothelioma. Nat Commun 2024; 15:7187. [PMID: 39168966 PMCID: PMC11339264 DOI: 10.1038/s41467-024-49842-5] [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: 10/04/2023] [Accepted: 06/18/2024] [Indexed: 08/23/2024] Open
Abstract
Malignant mesothelioma is a rare tumour caused by asbestos exposure that originates mainly from the pleural lining or the peritoneum. Treatment options are limited, and the prognosis is dismal. Although immune checkpoint blockade (ICB) can improve survival outcomes, the determinants of responsiveness remain elusive. Here, we report the outcomes of a multi-centre phase II clinical trial (MiST4, NCT03654833) evaluating atezolizumab and bevacizumab (AtzBev) in patients with relapsed mesothelioma. We also use tumour tissue and gut microbiome sequencing, as well as tumour spatial immunophenotyping to identify factors associated with treatment response. MIST4 met its primary endpoint with 50% 12-week disease control, and the treatment was tolerable. Aneuploidy, notably uniparental disomy (UPD), homologous recombination deficiency (HRD), epithelial-mesenchymal transition and inflammation with CD68+ monocytes were identified as tumour-intrinsic resistance factors. The log-ratio of gut-resident microbial genera positively correlated with radiological response to AtzBev and CD8+ T cell infiltration, but was inversely correlated with UPD, HRD and tumour infiltration by CD68+ monocytes. In summary, a model is proposed in which both intrinsic and extrinsic determinants in mesothelioma cooperate to modify the tumour microenvironment and confer clinical sensitivity to AtzBev. Gut microbiota represent a potentially modifiable factor with potential to improve immunotherapy outcomes for individuals with this cancer of unmet need.
Collapse
Affiliation(s)
- Min Zhang
- National Institute for Health Research Biomedical Research Centre & Cancer Research UK Experimental Cancer Medicine Centre, University of Leicester, Leicester, UK
- Novogene Corporation, Beijing, China
| | - Aleksandra Bzura
- National Institute for Health Research Biomedical Research Centre & Cancer Research UK Experimental Cancer Medicine Centre, University of Leicester, Leicester, UK
| | - Essa Y Baitei
- National Institute for Health Research Biomedical Research Centre & Cancer Research UK Experimental Cancer Medicine Centre, University of Leicester, Leicester, UK
- Center for Genomic Medicine, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Zisen Zhou
- Department of Informatics, University of Leicester, Leicester, UK
| | - Jake B Spicer
- National Institute for Health Research Biomedical Research Centre & Cancer Research UK Experimental Cancer Medicine Centre, University of Leicester, Leicester, UK
| | - Charlotte Poile
- National Institute for Health Research Biomedical Research Centre & Cancer Research UK Experimental Cancer Medicine Centre, University of Leicester, Leicester, UK
| | - Jan Rogel
- National Institute for Health Research Biomedical Research Centre & Cancer Research UK Experimental Cancer Medicine Centre, University of Leicester, Leicester, UK
| | - Amy Branson
- Leicester Clinical Trials Unit, University of Leicester, Leicester, UK
| | - Amy King
- Department of Oncology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Shaun Barber
- Leicester Clinical Trials Unit, University of Leicester, Leicester, UK
| | - Tamihiro Kamata
- National Institute for Health Research Biomedical Research Centre & Cancer Research UK Experimental Cancer Medicine Centre, University of Leicester, Leicester, UK
| | - Joanna Dzialo
- National Institute for Health Research Biomedical Research Centre & Cancer Research UK Experimental Cancer Medicine Centre, University of Leicester, Leicester, UK
| | - James Harber
- Harry Perkins Institute of Medical Research and The University of Western Australia Centre for Cancer Research, Perth, WA, Australia
| | | | - Nada Nusrat
- National Institute for Health Research Biomedical Research Centre & Cancer Research UK Experimental Cancer Medicine Centre, University of Leicester, Leicester, UK
| | - Daniel Faulkner
- National Institute for Health Research Biomedical Research Centre & Cancer Research UK Experimental Cancer Medicine Centre, University of Leicester, Leicester, UK
| | | | - Luke Nolan
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Jens C Hahne
- National Institute for Health Research Biomedical Research Centre & Cancer Research UK Experimental Cancer Medicine Centre, University of Leicester, Leicester, UK
| | - Molly Scotland
- Department of Oncology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Harriet Walter
- National Institute for Health Research Biomedical Research Centre & Cancer Research UK Experimental Cancer Medicine Centre, University of Leicester, Leicester, UK
- Department of Oncology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | | | - Bruno Morgan
- Department of Radiology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Amrita Bajaj
- Department of Radiology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Cassandra Brookes
- Leicester Clinical Trials Unit, University of Leicester, Leicester, UK
| | - Edward J Hollox
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Dominika Lubawska
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Maymun Jama
- National Institute for Health Research Biomedical Research Centre & Cancer Research UK Experimental Cancer Medicine Centre, University of Leicester, Leicester, UK
| | | | - Apostolos Nakas
- Department of Cardiothoracic Surgery, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Kudzayi Kutywayo
- Department of Cardiothoracic Surgery, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Jin-Li Luo
- Bioinformatics and Statistics Analysis Hub, University of Leicester, Leicester, UK
| | | | - Andrea Cooper
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Koirobi Halder
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Peter Wells-Jordan
- National Institute for Health Research Biomedical Research Centre & Cancer Research UK Experimental Cancer Medicine Centre, University of Leicester, Leicester, UK
| | - Huiyu Zhou
- Department of Informatics, University of Leicester, Leicester, UK
| | - Frank Dudbridge
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Anne Thomas
- National Institute for Health Research Biomedical Research Centre & Cancer Research UK Experimental Cancer Medicine Centre, University of Leicester, Leicester, UK
- Department of Oncology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | | | - Catrin Pritchard
- National Institute for Health Research Biomedical Research Centre & Cancer Research UK Experimental Cancer Medicine Centre, University of Leicester, Leicester, UK
| | - Hongji Yang
- Department of Informatics, University of Leicester, Leicester, UK
| | - Michael Barer
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Dean A Fennell
- National Institute for Health Research Biomedical Research Centre & Cancer Research UK Experimental Cancer Medicine Centre, University of Leicester, Leicester, UK.
- Department of Oncology, University Hospitals of Leicester NHS Trust, Leicester, UK.
| |
Collapse
|
136
|
Najar TA, Hao Y, Hao Y, Romero-Meza G, Dolynuk A, Littman DR. Microbiota-induced plastic T cells enhance immune control of antigen-sharing tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607605. [PMID: 39185202 PMCID: PMC11343098 DOI: 10.1101/2024.08.12.607605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Therapies that harness the immune system to target and eliminate tumor cells have revolutionized cancer care. Immune checkpoint blockade (ICB), which boosts the anti-tumor immune response by inhibiting negative regulators of T cell activation1-3, is remarkably successful in a subset of cancer patients, yet a significant proportion do not respond to treatment, emphasizing the need to understand factors influencing the therapeutic efficacy of ICB4-9. The gut microbiota, consisting of trillions of microorganisms residing in the gastrointestinal tract, has emerged as a critical determinant of immune function and response to cancer immunotherapy, with multiple studies demonstrating association of microbiota composition with clinical response10-16. However, a mechanistic understanding of how gut commensal bacteria influence the efficacy of ICB remains elusive. Here we utilized a gut commensal microorganism, segmented filamentous bacteria (SFB), which induces an antigen-specific Th17 cell effector program17, to investigate how colonization with it affects the efficacy of ICB in restraining distal growth of tumors sharing antigen with SFB. We find that anti-PD-1 treatment effectively inhibits the growth of implanted SFB antigen-expressing melanoma only if mice are colonized with SFB. Through T cell receptor clonal lineage tracing, fate mapping, and peptide-MHC tetramer staining, we identify tumor-associated SFB-specific Th1-like cells derived from the homeostatic Th17 cells induced by SFB colonization in the small intestine lamina propria. These gut-educated ex-Th17 cells produce high levels of the pro-inflammatory cytokines IFN-γ and TNF-α, and promote expansion and effector functions of CD8+ tumor-infiltrating cytotoxic lymphocytes, thereby controlling tumor growth. A better understanding of how distinct intestinal commensal microbes can promote T cell plasticity-dependent responses against antigen-sharing tumors may allow for the design of novel cancer immunotherapeutic strategies.
Collapse
Affiliation(s)
- Tariq A. Najar
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Yuan Hao
- Applied Bioinformatics Laboratories, Division of Advanced Research Technologies, New York, New York University School of Medicine, New York, NY 10016, USA
- Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Yuhan Hao
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
- New York Genome Center, New York, NY 10013, USA
| | - Gabriela Romero-Meza
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
- Howard Hughes Medical Institute, New York, NY 10016, USA
| | - Alexandra Dolynuk
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
- Howard Hughes Medical Institute, New York, NY 10016, USA
| | - Dan R. Littman
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
- Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
- Howard Hughes Medical Institute, New York, NY 10016, USA
| |
Collapse
|
137
|
Su X, Li J, Xu X, Ye Y, Wang C, Pang G, Liu W, Liu A, Zhao C, Hao X. Strategies to enhance the therapeutic efficacy of anti-PD-1 antibody, anti-PD-L1 antibody and anti-CTLA-4 antibody in cancer therapy. J Transl Med 2024; 22:751. [PMID: 39123227 PMCID: PMC11316358 DOI: 10.1186/s12967-024-05552-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Although immune checkpoint inhibitors (anti-PD-1 antibody, anti-PD-L1 antibody, and anti-CTLA-4 antibody) have displayed considerable success in the treatment of malignant tumors, the therapeutic effect is still unsatisfactory for a portion of patients. Therefore, it is imperative to develop strategies to enhance the effect of these ICIs. Increasing evidence strongly suggests that the key to this issue is to transform the tumor immune microenvironment from a state of no or low immune infiltration to a state of high immune infiltration and enhance the tumor cell-killing effect of T cells. Therefore, some combination strategies have been proposed and this review appraise a summary of 39 strategies aiming at enhancing the effectiveness of ICIs, which comprise combining 10 clinical approaches and 29 foundational research strategies. Moreover, this review improves the comprehensive understanding of combination therapy with ICIs and inspires novel ideas for tumor immunotherapy.
Collapse
Affiliation(s)
- Xin Su
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Jian Li
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Xiao Xu
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Youbao Ye
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Cailiu Wang
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Guanglong Pang
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Wenxiu Liu
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Ang Liu
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Changchun Zhao
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Xiangyong Hao
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China.
| |
Collapse
|
138
|
Zhu Y, Liu W, Wang M, Wang X, Wang S. Causal roles of skin microbiota in skin cancers suggested by genetic study. Front Microbiol 2024; 15:1426807. [PMID: 39161599 PMCID: PMC11330880 DOI: 10.3389/fmicb.2024.1426807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/19/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND There is evidence from observational studies that skin microbiota is linked to skin cancers. Nevertheless, the causal association between skin microbiota and skin cancers is yet to be fully clarified. METHODS A bidirectional two-sample Mendelian randomization (MR) was performed to determine the causal relationship between skin microbiota and skin cancers. A total of 294 skin microbial taxa were identified from the first genome-wide association study across three skin microenvironments of two German population cohorts. Summary data of three skin cancers (malignant melanoma, squamous cell carcinoma, and basal cell carcinoma) were obtained from the FinnGen consortium. Moreover, sensitivity analysis examined horizontal pleiotropy and heterogeneity, and microenvironment-based meta-analysis confirmed the reliability of the results. RESULTS We identified 65 nominal causalities and 5 strong causal associations between skin microbiota and skin cancers. Among them, the class Bacilli revealed a bidirectional positive relationship with malignant melanoma. The class Betaproteobacteria and class Gammaproteobacteria demonstrated a causal association with an elevated risk of malignant melanoma and basal cell carcinoma, respectively. In the reverse MR analysis, malignant melanoma was associated with a lower abundance of phylum Bacteroidetes. There were no indications of significant heterogeneity in instrumental variables or evidence of horizontal pleiotropy. CONCLUSION Our MR analysis indicated bidirectional causal associations between skin microbiota and skin cancers, and had the potential to offer novel perspectives on the mechanistic of microbiota-facilitated carcinogenesis.
Collapse
Affiliation(s)
- Yuhang Zhu
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Wanguo Liu
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Mei Wang
- Department of Dermatology, The First Hospital of Jilin University, Changchun, China
| | - Xu Wang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Sibo Wang
- Department of Neurology, Center for Neuroscience, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
139
|
Sun L, Ying J, Guo R, Jia L, Zhang H. A bibliometric analysis of global research on microbial immune microenvironment in melanoma from 2012 to 2022. Skin Res Technol 2024; 30:e70017. [PMID: 39167029 PMCID: PMC11337907 DOI: 10.1111/srt.70017] [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: 07/03/2024] [Accepted: 07/20/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Melanoma is an aggressive malignancy primarily impacting the skin, mucous membranes, and pigment epithelium. The tumor microbial microenvironment encompasses both the microorganisms inhabiting the tumor vicinity and the environmental factors influencing their interactions. Emerging evidence highlights the pivotal role of the microbial immune microenvironment in melanoma. METHODS We conducted an extensive review of scholarly works published from 2012 to 2022, utilizing The Web of Science Core Collection. Subsequently, we employed analytical tools such as VOSviewer, CiteSpace, and the R programming language to scrutinize prevailing research patterns within this domain. RESULTS A sum of 513 articles were pinpointed, with notable input coming from the United States and China. Harvard University stood out as the top-contributing institution, while the journal Science received the most citations. Current research within this sphere chiefly focuses on two principal domains: the gut microbiota and the PD-L1 pathway concerning melanoma treatment. CONCLUSION The study offers an extensive analysis and overview of the worldwide research landscape concerning the immune microenvironment with a focus on microbes in melanoma. It underscores the promising prospects for harnessing the microbial immune microenvironment's potential in melanoma. These findings furnish valuable insights and guidance for advancing scientific inquiry and refining clinical approaches within this dynamic field.
Collapse
Affiliation(s)
- Lin Sun
- Department of RadiotherapyYantaishan HospitalShandongChina
| | - Jianghui Ying
- Department of Plastic SurgeryShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Rong Guo
- Department of Plastic SurgeryShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Lingling Jia
- Department of Plastic SurgeryShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Hongyi Zhang
- Department of Plastic SurgeryShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| |
Collapse
|
140
|
Taylor MM, Nelson KC, Dimitriou F. Skin Cancer Precursors: From Cancer Genomics to Early Diagnosis. Hematol Oncol Clin North Am 2024; 38:851-868. [PMID: 38782646 DOI: 10.1016/j.hoc.2024.04.005] [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] [Indexed: 05/25/2024]
Abstract
Skin cancers, including melanoma and keratinocyte carcinomas, are responsible for increasing health care burden internationally. Risk stratification and early detection are paramount for prevention and less risky treatment to overall improve patient outcomes and disease morbidity. Here, the authors discuss the key concepts leading to skin cancer initiation and progression. The authors also outline precursor and progression models for melanoma and keratinocyte carcinomas, including discussion of genetic alterations associated with the various stages of progression. Finally, the authors discuss the significance of immunoediting and the drivers behind increased risk of cutaneous malignancy in the state of immune dysregulation.
Collapse
Affiliation(s)
- Madison M Taylor
- John P. and Kathrine G. McGovern Medical School, The University of Texas Health Science Center, 6431 Fannin Street, Houston, TX 77030, USA; Department of Dermatology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1452, Houston, TX 77030, USA
| | - Kelly C Nelson
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1452, Houston, TX 77030, USA.
| | - Florentia Dimitriou
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX 77030, USA; Department of Dermatology, University Hospital of Zurich, University of Zurich, Rämistrasse 100, 8091 Zürich, Switzerland
| |
Collapse
|
141
|
Espelage L, Wagner N, Placke JM, Ugurel S, Tasdogan A. The Interplay between Metabolic Adaptations and Diet in Cancer Immunotherapy. Clin Cancer Res 2024; 30:3117-3127. [PMID: 38771898 DOI: 10.1158/1078-0432.ccr-22-3468] [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: 07/06/2023] [Revised: 11/07/2023] [Accepted: 04/15/2024] [Indexed: 05/23/2024]
Abstract
Over the past decade, cancer immunotherapy has significantly advanced through the introduction of immune checkpoint inhibitors and the augmentation of adoptive cell transfer to enhance the innate cancer defense mechanisms. Despite these remarkable achievements, some cancers exhibit resistance to immunotherapy, with limited patient responsiveness and development of therapy resistance. Metabolic adaptations in both immune cells and cancer cells have emerged as central contributors to immunotherapy resistance. In the last few years, new insights emphasized the critical role of cancer and immune cell metabolism in animal models and patients. During therapy, immune cells undergo important metabolic shifts crucial for their acquired effector function against cancer cells. However, cancer cell metabolic rewiring and nutrient competition within tumor microenvironment (TME) alters many immune functions, affecting their fitness, polarization, recruitment, and survival. These interactions have initiated the development of novel therapies targeting tumor cell metabolism and favoring antitumor immunity within the TME. Furthermore, there has been increasing interest in comprehending how diet impacts the response to immunotherapy, given the demonstrated immunomodulatory and antitumor activity of various nutrients. In conclusion, recent advances in preclinical and clinical studies have highlighted the capacity of immune-based cancer therapies. Therefore, further exploration into the metabolic requirements of immune cells within the TME holds significant promise for the development of innovative therapeutic approaches that can effectively combat cancer in patients.
Collapse
Affiliation(s)
- Lena Espelage
- Department of Dermatology, University Hospital Essen and German Cancer Consortium (DKTK), Essen, Germany
| | - Natalie Wagner
- Department of Dermatology, University Hospital Essen and German Cancer Consortium (DKTK), Essen, Germany
| | - Jan-Malte Placke
- Department of Dermatology, University Hospital Essen and German Cancer Consortium (DKTK), Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen and German Cancer Consortium (DKTK), Essen, Germany
| | - Alpaslan Tasdogan
- Department of Dermatology, University Hospital Essen and German Cancer Consortium (DKTK), Essen, Germany
| |
Collapse
|
142
|
Liu X, Li S, Wang L, Ma K. Microecological regulation in HCC therapy: Gut microbiome enhances ICI treatment. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167230. [PMID: 38734322 DOI: 10.1016/j.bbadis.2024.167230] [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] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 05/13/2024]
Abstract
The exploration of the complex mechanisms of cancer immunotherapy is rapidly evolving worldwide, and our focus is on the interaction of hepatocellular carcinoma (HCC) with immune checkpoint inhibitors (ICIs), particularly as it relates to the regulatory role of the gut microbiome. An important basis for the induction of immune responses in HCC is the presence of specific anti-tumor cells that can be activated and reinforced by ICIs, which is why the application of ICIs results in sustained tumor response rates in the majority of HCC patients. However, mechanisms of acquired resistance to immunotherapy in unresectable HCC result in no long-term benefit for some patients. The significant heterogeneity of inter-individual differences in the gut microbiome in response to treatment with ICIs makes it possible to target modulation of specific gut microbes to assist in augmenting checkpoint blockade therapies in HCC. This review focuses on the complex relationship between the gut microbiome, host immunity, and HCC, and emphasizes that manipulating the gut microbiome to improve response rates to cancer ICI therapy is a clinical strategy with unlimited potential.
Collapse
Affiliation(s)
- Xuliang Liu
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Shiyao Li
- Department of Respiratory Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Liming Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China; Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China; Engineering Technology Research Center for Translational Medicine, The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China.
| | - Kexin Ma
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
143
|
Zhou L, Fan S, Zhang W, Wang D, Tang D. Microbes in the tumor microenvironment: New additions to break the tumor immunotherapy dilemma. Microbiol Res 2024; 285:127777. [PMID: 38797111 DOI: 10.1016/j.micres.2024.127777] [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: 12/11/2023] [Revised: 04/26/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024]
Abstract
Immunotherapies currently used in clinical practice are unsatisfactory in terms of therapeutic response and toxic side effects, and therefore new immunotherapies need to be explored. Intratumoral microbiota (ITM) exists in the tumor environment (TME) and reacts with its components. On the one hand, ITM promotes antigen delivery to tumor cells or provides cross-antigens to promote immune cells to attack tumors. On the other hand, ITM affects the activity of immune cells and stromal cells. We also summarize the dialog pathways by which ITM crosstalks with components within the TME, particularly the interferon pathway. This interaction between ITM and TME provides new ideas for tumor immunotherapy. By analyzing the bidirectional role of ITM in TME and combining it with its experimental and clinical status, we summarized the adjuvant role of ITM in immunotherapy. We explored the potential applications of using ITM as tumor immunotherapy, such as a healthy diet, fecal transplantation, targeted ITM, antibiotics, and probiotics, to provide a new perspective on the use of ITM in tumor immunotherapy.
Collapse
Affiliation(s)
- Lujia Zhou
- Clinical Medical college, Yangzhou University, Yangzhou, Jiangsu Province 225000, China.
| | - Shiying Fan
- Clinical Medical college, Yangzhou University, Yangzhou, Jiangsu Province 225000, China.
| | - Wenjie Zhang
- School of Medicine, Chongqing University, Chongqing 400030, China.
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, P. R. China.
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, P. R. China.
| |
Collapse
|
144
|
Xiang Y, Zhang C, Wang J, Cheng Y, Wang K, Wang L, Tong Y, Yan D. Role of blood metabolites in mediating the effect of gut microbiome on the mutated-RAS/BRAF metastatic colorectal cancer-specific survival. Int J Colorectal Dis 2024; 39:116. [PMID: 39046546 PMCID: PMC11269474 DOI: 10.1007/s00384-024-04686-9] [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] [Accepted: 07/09/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Recent studies have linked alterations in the gut microbiome and metabolic disruptions to the invasive behavior and metastasis of colorectal cancer (CRC), thus affecting patient prognosis. However, the specific relationship among gut microbiome, metabolite profiles, and mutated-RAS/BRAF metastatic colorectal cancer (M-mCRC) remains unclear. Furthermore, the potential mechanisms and prognostic implications of metabolic changes induced by gut microbiome alterations in patients with M-mCRC still need to be better understood. METHODS We conducted Mendelian randomization (MR) to evaluate the causal relationship of genetically predicted 196 gut microbiome features and 1400 plasma metabolites/metabolite ratios on M-mCRC-specific survival. Additionally, we identified significant gut microbiome-metabolites/metabolite ratio associations based on M-mCRC. Metabolite information was annotated, and functional annotation and pathway enrichment analyses were performed on shared proteins corresponding to significant metabolite ratios, aiming to reveal potential mechanisms by which gut microbiome influences M-mCRC prognosis via modulation of human metabolism. RESULTS We identified 11 gut microbiome features and 49 known metabolites/metabolite ratios correlated with M-mCRC-specific survival. Furthermore, we identified 17 gut microbiome-metabolite/metabolite ratio associations specific to M-mCRC, involving eight lipid metabolites and three bilirubin degradation products. The shared proteins corresponding to significant metabolite ratios were predominantly localized within the integral component of the membrane and exhibited enzymatic activities such as glucuronosyltransferase and UDP-glucuronosyltransferase, crucial in processes such as glucuronidation, bile secretion, and lipid metabolism. Moreover, these proteins were significantly enriched in pathways related to ascorbate and aldarate metabolism, pentose and glucuronate interconversions, steroid hormone biosynthesis, and bile secretion. CONCLUSION Our study offers novel insights into the potential mechanisms underlying the impact of the gut microbiome on the prognosis of M-mCRC. These findings serve as a meaningful reference for exploring potential therapeutic targets and strategies in the future.
Collapse
Affiliation(s)
- Yaoxian Xiang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Chan Zhang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Jing Wang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Yurong Cheng
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Kangjie Wang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Li Wang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Yingying Tong
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Dong Yan
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China.
| |
Collapse
|
145
|
Meza-Perez S, Liu M, Silva-Sanchez A, Morrow CD, Eipers PG, Lefkowitz EJ, Ptacek T, Scharer CD, Rosenberg AF, Hill DD, Arend RC, Gray MJ, Randall TD. Proteobacteria impair anti-tumor immunity in the omentum by consuming arginine. Cell Host Microbe 2024; 32:1177-1191.e7. [PMID: 38942027 PMCID: PMC11245731 DOI: 10.1016/j.chom.2024.06.003] [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: 11/16/2023] [Revised: 04/19/2024] [Accepted: 06/04/2024] [Indexed: 06/30/2024]
Abstract
Gut microbiota influence anti-tumor immunity, often by producing immune-modulating metabolites. However, microbes consume a variety of metabolites that may also impact host immune responses. We show that tumors grow unchecked in the omenta of microbe-replete mice due to immunosuppressive Tregs. By contrast, omental tumors in germ-free, neomycin-treated mice or mice colonized with altered Schaedler's flora (ASF) are spontaneously eliminated by CD8+ T cells. These mice lack Proteobacteria capable of arginine catabolism, causing increases in serum arginine that activate the mammalian target of the rapamycin (mTOR) pathway in Tregs to reduce their suppressive capacity. Transfer of the Proteobacteria, Escherichia coli (E. coli), but not a mutant unable to catabolize arginine, to ASF mice reduces arginine levels, restores Treg suppression, and prevents tumor clearance. Supplementary arginine similarly decreases Treg suppressive capacity, increases CD8+ T cell effectiveness, and reduces tumor burden. Thus, microbial consumption of arginine alters anti-tumor immunity, offering potential therapeutic strategies for tumors in visceral adipose tissue.
Collapse
Affiliation(s)
- Selene Meza-Perez
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Mingyong Liu
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Aaron Silva-Sanchez
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Casey D Morrow
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Peter G Eipers
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Elliot J Lefkowitz
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Travis Ptacek
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Alexander F Rosenberg
- Department of Biomedical Informatics and Data Science, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Dave D Hill
- Department of Biomedical Informatics and Data Science, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Rebecca C Arend
- Department of Obstetrics and Gynecology, Division of Gynecological Oncology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michael J Gray
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Troy D Randall
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
146
|
Drew L. Faecal transplants can treat some cancers - but probably won't ever be widely used. Nature 2024:10.1038/d41586-024-02212-z. [PMID: 38965449 DOI: 10.1038/d41586-024-02212-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
|
147
|
Zhang Y, Zhao X, Zhang J, Zhang Y, Wei Y. Advancements in the impact of human microbiota and probiotics on leukemia. Front Microbiol 2024; 15:1423838. [PMID: 39021626 PMCID: PMC11251910 DOI: 10.3389/fmicb.2024.1423838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024] Open
Abstract
The human gut microbiota is a complex ecosystem that plays a crucial role in promoting the interaction between the body and its environment. It has been increasingly recognized that the gut microbiota has diverse physiological functions. Recent studies have shown a close association between the gut microbiota and the development of certain tumors, including leukemia. Leukemia is a malignant clonal disease characterized by the uncontrolled growth of one or more types of blood cells, which is the most common cancer in children. The imbalance of gut microbiota is linked to the pathological mechanisms of leukemia. Probiotics, which are beneficial microorganisms that help maintain the balance of the host microbiome, play a role in regulating gut microbiota. Probiotics have the potential to assist in the treatment of leukemia and improve the clinical prognosis of leukemia patients. This study reviews the relationship between gut microbiota, probiotics, and the progression of leukemia based on current research. In addition, utilizing zebrafish leukemia models in future studies might reveal the specific mechanisms of their interactions, thereby providing new insights into the clinical treatment of leukemia. In conclusion, further investigation is still needed to fully understand the accurate role of microbes in leukemia.
Collapse
Affiliation(s)
| | | | | | - Yaodong Zhang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, School of Pharmaceutical Sciences, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital Zhengzhou Children’s Hospital, Zhengzhou University, Zhengzhou, China
| | - Yongjun Wei
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, School of Pharmaceutical Sciences, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital Zhengzhou Children’s Hospital, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
148
|
Cho YS, Han K, Xu J, Moon JJ. Novel strategies for modulating the gut microbiome for cancer therapy. Adv Drug Deliv Rev 2024; 210:115332. [PMID: 38759702 PMCID: PMC11268941 DOI: 10.1016/j.addr.2024.115332] [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: 01/29/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
Recent advancements in genomics, transcriptomics, and metabolomics have significantly advanced our understanding of the human gut microbiome and its impact on the efficacy and toxicity of anti-cancer therapeutics, including chemotherapy, immunotherapy, and radiotherapy. In particular, prebiotics, probiotics, and postbiotics are recognized for their unique properties in modulating the gut microbiota, maintaining the intestinal barrier, and regulating immune cells, thus emerging as new cancer treatment modalities. However, clinical translation of microbiome-based therapy is still in its early stages, facing challenges to overcome physicochemical and biological barriers of the gastrointestinal tract, enhance target-specific delivery, and improve drug bioavailability. This review aims to highlight the impact of prebiotics, probiotics, and postbiotics on the gut microbiome and their efficacy as cancer treatment modalities. Additionally, we summarize recent innovative engineering strategies designed to overcome challenges associated with oral administration of anti-cancer treatments. Moreover, we will explore the potential benefits of engineered gut microbiome-modulating approaches in ameliorating the side effects of immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Young Seok Cho
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kai Han
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 21009, China; Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 21009, China
| | - Jin Xu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
149
|
Nakatsu G, Andreeva N, MacDonald MH, Garrett WS. Interactions between diet and gut microbiota in cancer. Nat Microbiol 2024; 9:1644-1654. [PMID: 38907007 DOI: 10.1038/s41564-024-01736-4] [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: 02/13/2023] [Accepted: 05/20/2024] [Indexed: 06/23/2024]
Abstract
Dietary patterns and specific dietary components, in concert with the gut microbiota, can jointly shape susceptibility, resistance and therapeutic response to cancer. Which diet-microbial interactions contribute to or mitigate carcinogenesis and how they work are important questions in this growing field. Here we interpret studies of diet-microbial interactions to assess dietary determinants of intestinal colonization by opportunistic and oncogenic bacteria. We explore how diet-induced expansion of specific gut bacteria might drive colonic epithelial tumorigenesis or create immuno-permissive tumour milieus and introduce recent findings that provide insight into these processes. Additionally, we describe available preclinical models that are widely used to study diet, microbiome and cancer interactions. Given the rising clinical interest in dietary modulations in cancer treatment, we highlight promising clinical trials that describe the effects of different dietary alterations on the microbiome and cancer outcomes.
Collapse
Affiliation(s)
- Geicho Nakatsu
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Natalia Andreeva
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Meghan H MacDonald
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Wendy S Garrett
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Harvard Chan Microbiome in Public Health Center, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
150
|
Bredon M, Danne C, Pham HP, Ruffié P, Bessede A, Rolhion N, Creusot L, Brot L, Alonso I, Langella P, Derosa L, Cortot AB, Routy B, Zitvogel L, Segata N, Sokol H. Faecalibaterium prausnitzii strain EXL01 boosts efficacy of immune checkpoint inhibitors. Oncoimmunology 2024; 13:2374954. [PMID: 38957477 PMCID: PMC11218805 DOI: 10.1080/2162402x.2024.2374954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024] Open
Abstract
Gut microbiota impacts responses to immune checkpoint inhibitors (ICI). A high level of Faecalibacterium prausnitzii have been associated with a positive response to ICI in multiple cancer types. Here, based on fecal shotgun metagenomics data, we show in two independent cohorts of patients with non-small cell lung cancer and advanced melanoma that a high level of F. prausnitzii at baseline is positively associated with a better clinical response to ICI. In MCA205 tumor-bearing mice, administration of F. prausnitzii strain EXL01, already in clinical development for Inflammatory Bowel Disease, restores the anti-tumor response to ICI in the context of antibiotic-induced microbiota perturbation at clinical and tumor transcriptomics level. In vitro, EXL01 strain enhances T cell activation in the presence of ICI. Interestingly, oral administration of EXL01 strain did not induce any change in fecal microbiota diversity or composition, suggesting a direct effect on immune cells in the small intestine. F. prausnitzii strain EXL01 will be evaluated as an adjuvant to ICI in multiple cancers in the near future.
Collapse
Affiliation(s)
- Marius Bredon
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Camille Danne
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | | | | | | | - Nathalie Rolhion
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Laura Creusot
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Loic Brot
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Iria Alonso
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Philippe Langella
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, Jouy-en-Josas, France
| | - Lisa Derosa
- UMR1015 Gustave Roussy Cancer Campus, Villejuif, France
- Département de Médecine Oncologique, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicetre, France
| | - Alexis B. Cortot
- Université de Lille, CHU Lille, Thoracic Oncology Department, Centre National de la Recherche Scientifique, INSERM, Institut Pasteur de Lille, UMR9020-UMR-S 1277-Canther, Lille, France
| | - Bertrand Routy
- Department of Medicine, Hematology-Oncology Division, University of Montreal Healthcare Centre (CHUM), Montreal, QC, Canada
- University of Montreal Research Center (CRCHUM), Montreal, QC, Canada
| | - Laurence Zitvogel
- UMR1015 Gustave Roussy Cancer Campus, Villejuif, France
- Département de Médecine Oncologique, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicetre, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Harry Sokol
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, Jouy-en-Josas, France
| |
Collapse
|