1
|
Qin L, Hu C, Zhao Q, Wang Y, Fan D, Lin A, Xiang L, Chen Y, Shao J. Unraveling the role of Ctla-4 in intestinal immune homeostasis through a novel Zebrafish model of inflammatory bowel disease. eLife 2025; 13:RP101932. [PMID: 40392591 PMCID: PMC12092003 DOI: 10.7554/elife.101932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic and relapsing immune-mediated disorder characterized by intestinal inflammation and epithelial injury. The underlying causes of IBD are not fully understood, but genetic factors have been implicated in genome-wide association studies, including CTLA-4, an essential negative regulator of T cell activation. However, establishing a direct link between CTLA-4 and IBD has been challenging due to the early lethality of CTLA-4 knockout mice. In this study, we identified zebrafish Ctla-4 homolog and investigated its role in maintaining intestinal immune homeostasis by generating a Ctla-4-deficient (ctla-4-/-) zebrafish line. These mutant zebrafish exhibited reduced weight, along with impaired epithelial barrier integrity and lymphocytic infiltration in their intestines. Transcriptomics analysis revealed upregulation of inflammation-related genes, disturbing immune system homeostasis. Moreover, single-cell RNA-sequencing analysis indicated increased Th2 cells and interleukin 13 expression, along with decreased innate lymphoid cells and upregulated proinflammatory cytokines. Additionally, Ctla-4-deficient zebrafish exhibited reduced diversity and an altered composition of the intestinal microbiota. All these phenotypes closely resemble those found in mammalian IBD. Lastly, supplementation with Ctla-4-Ig successfully alleviated intestinal inflammation in these mutants. Altogether, our findings demonstrate the pivotal role of Ctla-4 in maintaining intestinal homeostasis. Additionally, they offer substantial evidence linking CTLA-4 to IBD and establish a novel zebrafish model for investigating both the pathogenesis and potential treatments.
Collapse
Affiliation(s)
- Lulu Qin
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Chongbin Hu
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Qiong Zhao
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Yong Wang
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Dongdong Fan
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Aifu Lin
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Lixin Xiang
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Ye Chen
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
- Department of Genetic and Metabolic Disease, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouChina
| | - Jianzhong Shao
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and TechnologyQingdaoChina
| |
Collapse
|
2
|
Gao YQ, Tan YJ, Fang JY. Roles of the gut microbiota in immune-related adverse events: mechanisms and therapeutic intervention. Nat Rev Clin Oncol 2025:10.1038/s41571-025-01026-w. [PMID: 40369317 DOI: 10.1038/s41571-025-01026-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2025] [Indexed: 05/16/2025]
Abstract
Immune checkpoint inhibitors (ICIs) constitute a major breakthrough in the field of cancer therapy; their use has resulted in improved outcomes across various tumour types. However, ICIs can cause a diverse range of immune-related adverse events (irAEs) that present a considerable challenge to the efficacy and safety of these treatments. The gut microbiota has been demonstrated to have a crucial role in modulating the tumour immune microenvironment and thus influences the effectiveness of ICIs. Accumulating evidence indicates that alterations in the composition and function of the gut microbiota are also associated with an increased risk of irAEs, particularly ICI-induced colitis. Indeed, these changes in the gut microbiota can contribute to the pathogenesis of irAEs. In this Review, we first summarize the current clinical challenges posed by irAEs. We then focus on reported correlations between alterations in the gut microbiota and irAEs, especially ICI-induced colitis, and postulate mechanisms by which these microbial changes influence the occurrence of irAEs. Finally, we highlight the potential value of gut microbial changes as biomarkers for predicting irAEs and discuss gut microbial interventions that might serve as new strategies for the management of irAEs, including faecal microbiota transplantation, probiotic, prebiotic and/or postbiotic supplements, and dietary modulations.
Collapse
Affiliation(s)
- Ya-Qi Gao
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong-Jie Tan
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Rehermann B, Graham AL, Masopust D, Hamilton SE. Integrating natural commensals and pathogens into preclinical mouse models. Nat Rev Immunol 2025; 25:385-397. [PMID: 39562646 DOI: 10.1038/s41577-024-01108-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2024] [Indexed: 11/21/2024]
Abstract
Fundamental discoveries in many aspects of mammalian physiology have been made using laboratory mice as research models. These studies have been facilitated by the genetic tractability and inbreeding of such mice, the large set of immunological reagents that are available, and the establishment of environmentally controlled, high-throughput facilities. Such facilities typically include barriers to keep the mouse colonies free of pathogens and the frequent re-derivation of the mice severely limits their commensal flora. Because humans have co-evolved with microorganisms and are exposed to a variety of pathogens, a growing community of researchers posits that preclinical disease research can be improved by studying mice in the context of the microbiota and pathogens that they would encounter in the natural world. Here, we provide a perspective of how these different approaches can be combined and integrated to improve existing mouse models to enhance our understanding of disease mechanisms and develop new therapies for humans. We also propose that the term 'mice with natural microbiota' is more appropriate for describing these models than existing terms such as 'dirty mice'.
Collapse
Affiliation(s)
- Barbara Rehermann
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Andrea L Graham
- Department of Ecology & Evolutionary Biology, Princeton University, Princeton, NJ, USA
| | - David Masopust
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Sara E Hamilton
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
4
|
Cui X, Li C, Zhong J, Liu Y, Xiao P, Liu C, Zhao M, Yang W. Gut microbiota - bidirectional modulator: role in inflammatory bowel disease and colorectal cancer. Front Immunol 2025; 16:1523584. [PMID: 40370465 PMCID: PMC12075242 DOI: 10.3389/fimmu.2025.1523584] [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: 11/06/2024] [Accepted: 04/08/2025] [Indexed: 05/16/2025] Open
Abstract
The gut microbiota is a diverse ecosystem that significantly impacts human health and disease. This article focuses on how the gut microbiota interacts with inflammatory bowel diseases and colorectal tumors, especially through immune regulation. The gut microbiota plays a role in immune system development and regulation, while the body's immune status can also affect the composition of the microbiota. These microorganisms exert pathogenic effects or correct disease states in gastrointestinal diseases through the actions of toxins and secretions, inhibition of immune responses, DNA damage, regulation of gene expression, and protein synthesis. The microbiota and its metabolites are essential in the development and progression of inflammatory bowel diseases and colorectal tumors. The complexity and bidirectionality of this connection with tumors and inflammation might render it a new therapeutic target. Hence, we explore therapeutic strategies for the gut microbiota, highlighting the potential of probiotics and fecal microbiota transplantation to restore or adjust the microbial community. Additionally, we address the challenges and future research directions in this area concerning inflammatory bowel diseases and colorectal tumors.
Collapse
Affiliation(s)
- Xilun Cui
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jing Zhong
- Department of Medical Imaging, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Yuanda Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Pengtuo Xiao
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chang Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Mengwei Zhao
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Wei Yang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
5
|
Dong H, Peng Y, Wang X, Cui H. An updated review on immune checkpoint inhibitor-induced colitis: epidemiology, pathogenesis, treatment strategies, and the role of traditional Chinese medicine. Front Immunol 2025; 16:1551445. [PMID: 40165945 PMCID: PMC11955479 DOI: 10.3389/fimmu.2025.1551445] [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: 12/25/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
Immune checkpoint inhibitor-induced colitis (irColitis) is a common and severe adverse reaction to immune checkpoint inhibitors (ICIs), significantly impacting the treatment outcomes and quality of life of cancer patients. Epidemiological studies indicate that the incidence of irColitis is associated with factors such as the type of ICIs, the patient's gender, age, and medical history. Although the exact pathophysiology remains unclear, irColitis is thought to be related to immune system activation and dysregulation, gut microbiota imbalance, and impaired epithelial barrier function. This review summarized the epidemiology, clinical presentation, diagnostic criteria, and pathogenesis of irColitis. Additionally, the standard and novel therapeutic strategies of irColitis, including corticosteroids, biologics, and gut microbiota interventions, more importantly the potential and application of Traditional Chinese Medicine (TCM). Future researches call for deeper mechanistic investigations, the development of biomarkers, and reveal the integration of TCM therapies within individual immunotherapy frameworks.
Collapse
Affiliation(s)
- Huijing Dong
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Yanmei Peng
- Department of Oncology, Fangshan Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Xinmeng Wang
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Huijuan Cui
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
6
|
Chen YH, Zaldana K, Yeung F, Vujkovic-Cvijin I, Downie AE, Lin JD, Yang Y, Herrmann C, Oyesola O, Rozenberg F, Schwartz RE, Kim D, Tio K, Belkaid Y, Loke P, Graham AL, Koralov SB, Cadwell K. Rewilding catalyzes maturation of the humoral immune system. SCIENCE ADVANCES 2025; 11:eads2364. [PMID: 40053586 PMCID: PMC11887799 DOI: 10.1126/sciadv.ads2364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025]
Abstract
Inbred mice used for biomedical research display an underdeveloped immune system compared with adult humans, which is attributed in part to the artificial laboratory environment. Despite representing a central component of adaptive immunity, the impact of the laboratory environment on the B cell compartment has not been investigated in detail. Here, we performed an in-depth examination of B cells following rewilding, the controlled release of inbred laboratory mice into an outdoor enclosure. In rewilded mice, we observed B cells in circulation with increased signs of maturation, alongside heightened germinal center responses within secondary lymphoid organs. Rewilding also expanded B cells in the gut, which was accompanied by elevated systemic levels of immunoglobulin G (IgG) and IgM antibodies reactive to the microbiota. Our findings indicate that exposing laboratory mice to a more natural environment enhances B cell development to better reflect the immune system of free-living mammals.
Collapse
Affiliation(s)
- Ying-Han Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Kimberly Zaldana
- Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY 10016, USA
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Frank Yeung
- Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ivan Vujkovic-Cvijin
- Department of Biomedical Sciences & F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Alexander E. Downie
- Department of Primate Behavior and Evolution, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Jian-Da Lin
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei City, Taiwan
| | - Yi Yang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Christin Herrmann
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Oyebola Oyesola
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Felix Rozenberg
- SUNY Downstate Health Sciences University, New York, NY 11203, USA
| | - Robert E. Schwartz
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - David Kim
- Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Kurt Tio
- Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Yasmine Belkaid
- Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Metaorganism Laboratory, Department of Immunology, Pasteur Institute, Paris, France
| | - P’ng Loke
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrea L. Graham
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ 08544, USA
- Santa Fe Institute, Santa Fe, NM 87501, USA
| | - Sergei B. Koralov
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ken Cadwell
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
7
|
Wu X, Wei J, Ran W, Liu D, Yi Y, Gong M, Liu X, Gong Q, Li H, Gao J. The Gut Microbiota-Xanthurenic Acid-Aromatic Hydrocarbon Receptor Axis Mediates the Anticolitic Effects of Trilobatin. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412234. [PMID: 39836604 PMCID: PMC11904984 DOI: 10.1002/advs.202412234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/16/2024] [Indexed: 01/23/2025]
Abstract
Current treatments for ulcerative colitis (UC) remain limited, highlighting the need for novel therapeutic strategies. Trilobatin (TLB), a naturally derived food additive, exhibits potential anti-inflammatory properties. In this study, a dextran sulfate sodium (DSS)-induced animal model is used to investigate the effects of TLB on UC. It is found TLB significantly alleviates DSS-induced UC in mice, as evidenced by a reduction in the disease activity index, an increase in colon length, improvement in histopathological lesions. Furthermore, TLB treatment results in a decrease in proinflammatory cytokines and an increase in anti-inflammatory cytokines. TLB mitigates UC by modulating the intestinal microbiota, particularly Akkermansia, which enhances tryptophan metabolism and upregulates the production of xanthurenic acid (XANA). To confirm the role of TLB-induced microbiota changes, experiments are performed with pseudogerm-free mice and fecal transplantation. It is also identified XANA as a key metabolite that mediates TLB's protective effects. Both TLB and XANA markedly activate the aromatic hydrocarbon receptor (AhR). Administration of an AhR antagonist abrogates their protective effects, thereby confirming the involvement of AhR in the underlying mechanism. In conclusion, the study reveals a novel mechanism through which TLB alleviates UC by correcting microbiota imbalances, regulating tryptophan metabolism, enhancing XANA production, and activating AhR.
Collapse
Affiliation(s)
- Xiaoyu Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationDepartment of PharmacologyKey Laboratory of Basic Pharmacology of Guizhou Province and School of PharmacyZunyi Medical UniversityZunyi563000China
- Faculty of Functional Food and WineShenyang Pharmaceutical UniversityShenyang110016China
| | - Jiajia Wei
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationDepartment of PharmacologyKey Laboratory of Basic Pharmacology of Guizhou Province and School of PharmacyZunyi Medical UniversityZunyi563000China
| | - Wang Ran
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationDepartment of PharmacologyKey Laboratory of Basic Pharmacology of Guizhou Province and School of PharmacyZunyi Medical UniversityZunyi563000China
| | - Dongjing Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationDepartment of PharmacologyKey Laboratory of Basic Pharmacology of Guizhou Province and School of PharmacyZunyi Medical UniversityZunyi563000China
| | - Yang Yi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationDepartment of PharmacologyKey Laboratory of Basic Pharmacology of Guizhou Province and School of PharmacyZunyi Medical UniversityZunyi563000China
| | - Miaoxian Gong
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationDepartment of PharmacologyKey Laboratory of Basic Pharmacology of Guizhou Province and School of PharmacyZunyi Medical UniversityZunyi563000China
| | - Xin Liu
- School of Traditional Chinese MedicineLiaoning University of Traditional Chinese MedicineShenyang110016China
| | - Qihai Gong
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationDepartment of PharmacologyKey Laboratory of Basic Pharmacology of Guizhou Province and School of PharmacyZunyi Medical UniversityZunyi563000China
| | - Haibo Li
- School of Traditional Chinese MedicineLiaoning University of Traditional Chinese MedicineShenyang110016China
| | - Jianmei Gao
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationDepartment of PharmacologyKey Laboratory of Basic Pharmacology of Guizhou Province and School of PharmacyZunyi Medical UniversityZunyi563000China
| |
Collapse
|
8
|
Verheijden RJ, van Eijs MJM, Paganelli FL, Viveen MC, Rogers MRC, Top J, May AM, van de Wijgert JHHM, Suijkerbuijk KPM. Gut microbiome and immune checkpoint inhibitor toxicity. Eur J Cancer 2025; 216:115221. [PMID: 39793444 DOI: 10.1016/j.ejca.2025.115221] [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: 12/13/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
BACKGROUND Multiple studies have suggested that gut microbiome may influence immune checkpoint inhibitor (ICI) efficacy, but its association with immune-related adverse events (irAEs) is less well studied. In this prospective cohort study, we assessed whether gut microbiome composition at start, or changes during ICI, are associated with severe irAEs. METHODS Stool samples of cancer patients treated with anti-PD-1 ± anti-CTLA-4 were analyzed using 16S rRNA gene sequencing and metagenomic shotgun sequencing. Differences in alpha and beta diversity between patients with and without severe irAE were assessed, as well as differential relative abundance (RA) of taxa, MetaCyc pathways, and seven prespecified literature-based bacterial groups including pathobionts and Ruminococcaceae. FINDINGS We analyzed 497 samples of 195 patients before and soon after starting ICI, at severe irAE onset and after starting immunosuppression. Mean RA of the pathobionts group was significantly higher in patients who developed a severe irAE (8.2 %) compared to those who did not (4.8 %; odds ratio 1.40; 95 %CI 1.07-1.87) at baseline, and also early during ICI treatment and at severe irAE onset. A significantly stronger decrease in RA of Ruminococcaceae after starting ICI was observed in patients who developed a severe irAE compared to those who did not. RAs of Ruminococcaceae, the genus Ruminococcus, and the species R. bromii and R. callidus were significantly lower at severe irAE onset compared to other time points. INTERPRETATION Gut microbiome dysbiosis signaled by higher RA of pathobionts and decrease in RA of Ruminococcaceae may predispose to severe irAEs.
Collapse
Affiliation(s)
- Rik J Verheijden
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands; Department of Epidemiology and Health Economics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands; Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands
| | - Mick J M van Eijs
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands; Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands
| | - Fernanda L Paganelli
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands; Winclove Probiotics, Amsterdam, 1033JS, the Netherlands
| | - Marco C Viveen
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands
| | - Malbert R C Rogers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands
| | - Janetta Top
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands
| | - Anne M May
- Department of Epidemiology and Health Economics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands
| | - Janneke H H M van de Wijgert
- Department of Epidemiology and Health Economics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands
| | - Karijn P M Suijkerbuijk
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands.
| |
Collapse
|
9
|
Mahadevan A, Yazdanpanah O, Patel V, Benjamin DJ, Kalebasty AR. Ophthalmologic toxicities of antineoplastic agents in genitourinary cancers: Mechanisms, management, and clinical implications. Curr Probl Cancer 2025; 54:101171. [PMID: 39708456 DOI: 10.1016/j.currproblcancer.2024.101171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/19/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024]
Abstract
Genitourinary cancers affect over 480,000 patients in the United States annually. While promising therapeutic modalities continue to emerge, notably immune checkpoint inhibitors, molecular targeted therapies, antibody-drug conjugates, and radioligand therapies, these treatments are associated with a spectrum of adverse side-effects, including ophthalmologic toxicities. In this review, we cover the most commonly used antineoplastic agents for the kidneys, bladder, urinary tracts, prostate, testis, and penis, detailing mechanism, indication, and recent trials supporting their use. For each category of antineoplastic therapy, we describe the epidemiology, management, and clinical presentation, of common ophthalmologic toxicities stemming from these agents. This review serves to augment awareness and recognition of possible ophthalmologic manifestations resulting from the use of antineoplastic agents in genitourinary malignancy. Early identification of these side effects can hasten ophthalmology referral and ultimately improve visual outcomes in patients experiencing medication-induced ocular toxicities.
Collapse
Affiliation(s)
- Aditya Mahadevan
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| | - Omid Yazdanpanah
- Division of Hematology/Oncology, University of California Irvine Health, Orange, CA, USA.
| | - Vivek Patel
- Department of Ophthalmology, University of California Irvine Health, Orange, CA, USA.
| | | | | |
Collapse
|
10
|
McDaniel Mims B, Furr KL, Enriquez J, Grisham MB. Improving bench-to-bedside translation for acute graft-versus-host disease models. Dis Model Mech 2025; 18:DMM052084. [PMID: 40019007 PMCID: PMC11892683 DOI: 10.1242/dmm.052084] [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] [Indexed: 03/01/2025] Open
Abstract
The transplantation of allogeneic hematopoietic stem cells is a potentially curative treatment for hematological malignancies, inherited blood disorders and immune deficiencies. Unfortunately, 30-50% of patients receiving allogeneic hematopoietic stem cells will develop a potentially life-threatening inflammatory disease called acute graft-versus-host disease (aGVHD). In patients with aGVHD, graft-associated T cells, which typically target the skin, intestinal tract and liver, can also damage the lungs and lymphoid tissue. Damage to lymphoid tissue creates prolonged immunodeficiency that markedly increases the risk of infections and bleeding, resulting in considerable morbidity and mortality. Although mouse models of aGVHD have been instrumental to our understanding of this condition's pathogenesis, translation of preclinical data into new and more effective treatments for human disease has been limited for reasons that remain to be fully understood. However, evidence suggests that factors associated with mouse models of aGVHD likely contribute to these unsatisfactory results. In this Review, we identify and discuss the specific factors inherent to mouse models of aGVHD that may limit the translation of preclinical data to patient treatment, and suggest how to improve the translatability of these models.
Collapse
Affiliation(s)
- Brianyell McDaniel Mims
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kathryn L. Furr
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79423, USA
| | - Josue Enriquez
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Matthew B. Grisham
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79423, USA
| |
Collapse
|
11
|
Jiang SS, Kang ZR, Chen YX, Fang JY. The gut microbiome modulate response to immunotherapy in cancer. SCIENCE CHINA. LIFE SCIENCES 2025; 68:381-396. [PMID: 39235561 DOI: 10.1007/s11427-023-2634-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 06/05/2024] [Indexed: 09/06/2024]
Abstract
Gut microbiota have been reported to play an important role in the occurrence and development of malignant tumors. Currently, clinical studies have identified specific gut microbiota and its metabolites associated with efficacy of immunotherapy in multiple types of cancers. Preclinical investigations have elucidated that gut microbiota modulate the antitumor immunity and affect the efficacy of cancer immunotherapy. Certain microbiota and its metabolites may favorably remodel the tumor microenvironment by engaging innate and/or adaptive immune cells. Understanding how the gut microbiome interacts with cancer immunotherapy opens new avenues for improving treatment strategies. Fecal microbial transplants, probiotics, dietary interventions, and other strategies targeting the microbiota have shown promise in preclinical studies to enhance the immunotherapy. Ongoing clinical trials are evaluating these approaches. This review presents the recent advancements in understanding the dynamic interplay among the host immunity, the microbiome, and cancer immunotherapy, as well as strategies for modulating the microbiome, with a view to translating into clinical applications.
Collapse
Affiliation(s)
- Shan-Shan Jiang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China
| | - Zi-Ran Kang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China
| | - Ying-Xuan Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China.
| |
Collapse
|
12
|
Tang Y, Cai Q, Tian Z, Chen W, Tang H. Crosstalk between Gut Microbiota and Cancer Immunotherapy: Present Investigations and Future Perspective. RESEARCH (WASHINGTON, D.C.) 2025; 8:0600. [PMID: 39850365 PMCID: PMC11754537 DOI: 10.34133/research.0600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 01/25/2025]
Abstract
Gut microbiota is crucial for protecting the homeostasis of immune locally and systemically, and its dysbiosis is essentially correlated to tumorigenesis, cancer progression, and refractoriness to cancer treatments, including the novel immunotherapy. Increasing evidence unravel the intricate role of gut microbiota in reshaping tumor microenvironment and affecting the efficacy and toxicities of immunotherapy, which shed more light on the future applications of gut microbiota in efficacious biomarker and combination treatment of immunotherapy. To better grasp the underlying crosstalk between gut microbiota and immunotherapy, more experimental and clinical trials are indispensable for the customized gut microbiota-based treatments in cancer patients undergoing immunotherapy.
Collapse
Affiliation(s)
- Yuhui Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qiaoting Cai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhi Tian
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Wenkuan Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
13
|
Jia D, Chen S. Commensal fungi, a force to be reckoned with. Cell Host Microbe 2025; 33:6-8. [PMID: 39788097 DOI: 10.1016/j.chom.2024.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 01/12/2025]
Abstract
Fungal symbionts play a key role in maintaining host homeostasis. In a recent issue of Nature, Liao et al. show that Kazachstania pintolopesii, a symbiotic fungus in mice, is shielded from the host immune system during homeostasis but induces type 2 immunity during mucus fluctuations.
Collapse
Affiliation(s)
- Dingjiacheng Jia
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province 310058, China.
| | - Shujie Chen
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province 310058, China; Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang Province 310058, China.
| |
Collapse
|
14
|
Bruno P, Schüler T, Rosshart SP. Born to be wild: utilizing natural microbiota for reliable biomedical research. Trends Immunol 2025; 46:17-28. [PMID: 39690004 DOI: 10.1016/j.it.2024.11.013] [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: 10/11/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024]
Abstract
Laboratory mice housed under specific pathogen-free (SPF) conditions are the standard model in biomedical research. However, experiments with a particular inbred mouse strain performed in different laboratories often yield inconsistent or conflicting data due to housing-specific variations in the composition and diversity of SPF microbiota. These variations affect immune and nonimmune cell functions, leading to systemic physiological changes. Consequently, microbiota-dependent inconsistencies have raised general doubts regarding the suitability of mice as model organisms. Since stability positively correlates with biological diversity, we postulate that increasing species diversity can improve microbiota stability and mouse physiology, enhancing robustness, reproducibility, and experimental validity. Similar to the generation of inbred mouse strains in the last century, we suggest a worldwide initiative to define a transplantable 'wild' microbiota that stably colonizes mice irrespective of housing conditions.
Collapse
Affiliation(s)
- Philipp Bruno
- Department of Microbiome Research, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany.
| | - Stephan P Rosshart
- Department of Microbiome Research, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany; Department of Medicine II, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany.
| |
Collapse
|
15
|
Taner HF, Gong W, Fitzsimonds ZR, Li Z, Wu Y, He Y, Okuyama K, Cheng W, Kuczura J, Rajesh S, Manousidaki A, Feng S, Lee M, Nör F, Lanzel E, Demehri S, Polverini PJ, Nör JE, Wang TD, Que J, Wen H, Xie Y, Moon JJ, Lei YL. SOX2-induced IL1α-mediated immune suppression drives epithelial dysplasia malignant transformation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.06.626475. [PMID: 39713429 PMCID: PMC11661103 DOI: 10.1101/2024.12.06.626475] [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: 12/24/2024]
Abstract
Squamous cell carcinomas (SCC) are often preceded by potentially malignant precursor lesions, most of which remain benign. The terminal exhaustion phenotypes of effector T-cells and the accumulation of myeloid-derived suppressor cells (MDSC) have been thoroughly characterized in established SCC. However, it is unclear what precancerous lesions harbor a bona fide high risk for malignant transformation and how precancerous epithelial dysplasia drives the immune system to the point of no return. Here we show that expression of SRY-box transcription factor 2 (SOX2) in precancerous lesions imparts an irreversible risk that recruits suppressive myeloid cells by promoting the release of CCL2. We developed a unique genetically engineered mouse model (GEMM) to recapitulate the malignant transformation of epithelial dysplasia to SCC in the oral mucosa with high histologic and phenotypic fidelity. Using a combination of longitudinal human specimens and the Sox2-GEMM, we found that the myeloid cells in precancerous epithelial dysplasia exhibit a distinctive dichotomous profile featuring high levels of IL-1α-SLC2A1 and low levels of type-I interferon (IFN-I) signatures, which occurs before SCC emerges histologically. Brief priming of myeloid cells with IL-1α desensitizes them to IFN-I agonists and makes myeloid-derived suppressor cells (MDSC) even more suppressive of T-cell activation. Mechanistically, IL-1 activation represses the expression of DHHC3/7 enzymes, which are responsible for the palmitoylation of stimulator of interferon genes (STING). Early blockade of IL1 signaling using pharmacologic and genetic approaches similarly reduces MDSC and SLC2A1high myeloid cells, suppresses epithelial dysplasia transformation, and extends survival. This work establishes a previously unrecognized SOX2-CCL2-IL1 pathway that leads to irreversible immune escape when precancerous epithelial lesions transform.
Collapse
Affiliation(s)
- Hülya F. Taner
- Oral Health Sciences DDS/PhD Program, University of Michigan School of Dentistry, Ann Arbor, MI 48105, USA
| | - Wang Gong
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Zackary R. Fitzsimonds
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Zaiye Li
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Yuesong Wu
- Department of Statistics and Probability, Michigan State University, East Lansing, MI 48824, USA
| | - Yumin He
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Kohei Okuyama
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Wanqing Cheng
- Graduate School of Biomedical Sciences, the University of Texas M.D. Anderson Cancer Center and UTHealth Houston, Houston, TX 77030, USA
| | - Jung Kuczura
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Sashider Rajesh
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48105, USA
| | - Andriana Manousidaki
- Department of Statistics and Probability, Michigan State University, East Lansing, MI 48824, USA
| | - Shuo Feng
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Miki Lee
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Felipe Nör
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48105, USA
| | - Emily Lanzel
- Department of Oral Pathology, Radiology, and Medicine, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
| | - Shadmehr Demehri
- Department of Dermatology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Peter J. Polverini
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48105, USA
| | - Jacques E. Nör
- Oral Health Sciences DDS/PhD Program, University of Michigan School of Dentistry, Ann Arbor, MI 48105, USA
- Department of Otolaryngology – Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Department of Cariology, Restorative Science and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48105, USA
| | - Thomas D. Wang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Jianwen Que
- Department of Medicine, Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA
| | - Haitao Wen
- Department of Microbial Infection and Immunity, Ohio State University College of Medicine, Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Yuying Xie
- Department of Statistics and Probability, Michigan State University, East Lansing, MI 48824, USA
- Department of Computational Mathematics Science and Engineering, Michigan State University, East Lansing, MI 48864, USA
| | - James J. Moon
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
- Department of Pharmaceutical Sciences, University of Michigan College of Pharmacy, Ann Arbor, MI 48105, USA
| | - Yu Leo Lei
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Oral Health Sciences DDS/PhD Program, University of Michigan School of Dentistry, Ann Arbor, MI 48105, USA
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Graduate School of Biomedical Sciences, the University of Texas M.D. Anderson Cancer Center and UTHealth Houston, Houston, TX 77030, USA
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| |
Collapse
|
16
|
Kim CW, Kim HJ, Lee HK. Microbiome dynamics in immune checkpoint blockade. Trends Endocrinol Metab 2024; 35:996-1005. [PMID: 38705760 DOI: 10.1016/j.tem.2024.04.013] [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: 01/17/2024] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 05/07/2024]
Abstract
Immune checkpoint blockade (ICB) is one of the leading immunotherapies, although a variable extent of resistance has been observed among patients and across cancer types. Among the efforts underway to overcome this challenge, the microbiome has emerged as a factor affecting the responsiveness and efficacy of ICB. Active research, facilitated by advances in sequencing techniques, is assessing the predominant influence of the intestinal microbiome, as well as the effects of the presence of an intratumoral microbiome. In this review, we describe recent findings from clinical trials, observational studies of human patients, and animal studies on the impact of the microbiome on the efficacy of ICB, highlighting the role of the intestinal and tumor microbiomes and the contribution of methodological advances in their study.
Collapse
Affiliation(s)
- Chae Won Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea; Life Science Institute, KAIST, Daejeon 34141, Republic of Korea
| | - Hyun-Jin Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea; Life Science Institute, KAIST, Daejeon 34141, Republic of Korea
| | - Heung Kyu Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
17
|
Jiménez-Andrade Y, Flesher JL, Park JM. Cancer Therapy-induced Dermatotoxicity as a Window to Understanding Skin Immunity. Hematol Oncol Clin North Am 2024; 38:1011-1025. [PMID: 38866636 PMCID: PMC11368641 DOI: 10.1016/j.hoc.2024.05.002] [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: 06/14/2024]
Abstract
Pruritus, rash, and various other forms of dermatotoxicity are the most frequent adverse events among patients with cancer receiving targeted molecular therapy and immunotherapy. Immune checkpoint inhibitors, macrophage-targeting agents, and epidermal growth factor receptor/MEK inhibitors not only exert antitumor effects but also interfere with molecular pathways essential for skin immune homeostasis. Studying cancer therapy-induced dermatotoxicity helps us identify molecular mechanisms governing skin immunity and deepen our understanding of human biology. This review summarizes new mechanistic insights emerging from the analysis of cutaneous adverse events and discusses knowledge gaps that remain to be closed by future research.
Collapse
Affiliation(s)
- Yanek Jiménez-Andrade
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Charlestown, MA 02129, USA
| | - Jessica L Flesher
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Charlestown, MA 02129, USA
| | - Jin Mo Park
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Charlestown, MA 02129, USA.
| |
Collapse
|
18
|
Recaldin T, Steinacher L, Gjeta B, Harter MF, Adam L, Kromer K, Mendes MP, Bellavista M, Nikolaev M, Lazzaroni G, Krese R, Kilik U, Popovic D, Stoll B, Gerard R, Bscheider M, Bickle M, Cabon L, Camp JG, Gjorevski N. Human organoids with an autologous tissue-resident immune compartment. Nature 2024; 633:165-173. [PMID: 39143209 PMCID: PMC11374719 DOI: 10.1038/s41586-024-07791-5] [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: 08/31/2023] [Accepted: 07/05/2024] [Indexed: 08/16/2024]
Abstract
The intimate relationship between the epithelium and immune system is crucial for maintaining tissue homeostasis, with perturbations therein linked to autoimmune disease and cancer1-3. Whereas stem cell-derived organoids are powerful models of epithelial function4, they lack tissue-resident immune cells that are essential for capturing organ-level processes. We describe human intestinal immuno-organoids (IIOs), formed through self-organization of epithelial organoids and autologous tissue-resident memory T (TRM) cells, a portion of which integrate within the epithelium and continuously survey the barrier. TRM cell migration and interaction with epithelial cells was orchestrated by TRM cell-enriched transcriptomic programs governing cell motility and adhesion. We combined IIOs and single-cell transcriptomics to investigate intestinal inflammation triggered by cancer-targeting biologics in patients. Inflammation was associated with the emergence of an activated population of CD8+ T cells that progressively acquired intraepithelial and cytotoxic features. The appearance of this effector population was preceded and potentiated by a T helper-1-like CD4+ population, which initially produced cytokines and subsequently became cytotoxic itself. As a system amenable to direct perturbation, IIOs allowed us to identify the Rho pathway as a new target for mitigation of immunotherapy-associated intestinal inflammation. Given that they recapitulate both the phenotypic outcomes and underlying interlineage immune interactions, IIOs can be used to study tissue-resident immune responses in the context of tumorigenesis and infectious and autoimmune diseases.
Collapse
Affiliation(s)
- Timothy Recaldin
- Roche Innovation Center Basel, Roche Pharma Research and Early Development, Basel, Switzerland
| | - Linda Steinacher
- Roche Innovation Center Basel, Roche Pharma Research and Early Development, Basel, Switzerland
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland
- Hannover Medical School, Institute of Immunology, Hannover, Germany
| | - Bruno Gjeta
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Marius F Harter
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland
- Gustave Roussy Cancer Campus, University Paris-Saclay, Paris, France
| | - Lukas Adam
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland
| | - Kristina Kromer
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland
| | - Marisa Pimentel Mendes
- Roche Innovation Center Basel, Roche Pharma Research and Early Development, Basel, Switzerland
| | - Marina Bellavista
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland
| | - Mikhail Nikolaev
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland
| | - Giacomo Lazzaroni
- Roche Innovation Center Basel, Roche Pharma Research and Early Development, Basel, Switzerland
| | - Rok Krese
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland
| | - Umut Kilik
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Doris Popovic
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland
| | - Bilgenaz Stoll
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland
| | - Régine Gerard
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland
| | - Michael Bscheider
- Roche Innovation Center Basel, Roche Pharma Research and Early Development, Basel, Switzerland
| | - Marc Bickle
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland
| | - Lauriane Cabon
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland.
| | - J Gray Camp
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland.
| | - Nikolche Gjorevski
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland.
| |
Collapse
|
19
|
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
|
20
|
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
|
21
|
He S, Zuo J, Lin Z. Mitigating gut immune adverse effects in CTLA-4 blockade for antitumor efficacy. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1539-1541. [PMID: 38578517 DOI: 10.1007/s11427-024-2550-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 02/07/2024] [Indexed: 04/06/2024]
Affiliation(s)
- Shijun He
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jianping Zuo
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zemin Lin
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
22
|
Yin Q, Ni J, Ying J. Potential mechanisms and targeting strategies of the gut microbiota in antitumor immunity and immunotherapy. Immun Inflamm Dis 2024; 12:e1263. [PMID: 39031507 PMCID: PMC11259004 DOI: 10.1002/iid3.1263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 03/24/2024] [Accepted: 04/18/2024] [Indexed: 07/22/2024] Open
Abstract
BACKGROUND Immunotherapies, notably immune checkpoints inhibitors that target programmed death 1/programmed death ligand 1(PD-1/PD-L1) and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), had profoundly changed the way advanced and metastatic cancers are treated and dramatically improved overall and progression-free survival. AIMS This review article aimed to explore the underlying molecular mechanisms by which the gut microbiota affects antitumor immunity and the efficacy of cancer immunotherapy. METHODS We summarized the latest knowledge supporting the associations among the gut microbiota, antitumor immunity, and immunotherapy. Moreover, we disscussed the therapeutic strategy for improving immunotherapy efficacy by modulating gut microbiota in cancer treatment. RESULTS The potential molecular mechanisms underlying these associations are explained in terms of four aspects: immunomodulation, molecular mimicry, mamps, and microbial metabolites. CONCLUSION The gut microbiota significantly impacts antitumor immunity and alters the effectiveness of cancer immunotherapy.
Collapse
Affiliation(s)
- Qian Yin
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital)HangzhouZhejiangChina
| | - Jiao‐jiao Ni
- Department of Hepato‐Pancreato‐Biliary & Gastric Medical OncologyZhejiang Cancer HospitalHangzhouChina
| | - Jie‐er Ying
- Department of Hepato‐Pancreato‐Biliary & Gastric Medical OncologyZhejiang Cancer HospitalHangzhouChina
| |
Collapse
|
23
|
Chen J, Yang L, Ma Y, Zhang Y. Recent advances in understanding the immune microenvironment in ovarian cancer. Front Immunol 2024; 15:1412328. [PMID: 38903506 PMCID: PMC11188340 DOI: 10.3389/fimmu.2024.1412328] [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: 04/04/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024] Open
Abstract
The occurrence of ovarian cancer (OC) is a major factor in women's mortality rates. Despite progress in medical treatments, like new drugs targeting homologous recombination deficiency, survival rates for OC patients are still not ideal. The tumor microenvironment (TME) includes cancer cells, fibroblasts linked to cancer (CAFs), immune-inflammatory cells, and the substances these cells secrete, along with non-cellular components in the extracellular matrix (ECM). First, the TME mainly plays a role in inhibiting tumor growth and protecting normal cell survival. As tumors progress, the TME gradually becomes a place to promote tumor cell progression. Immune cells in the TME have attracted much attention as targets for immunotherapy. Immune checkpoint inhibitor (ICI) therapy has the potential to regulate the TME, suppressing factors that facilitate tumor advancement, reactivating immune cells, managing tumor growth, and extending the survival of patients with advanced cancer. This review presents an outline of current studies on the distinct cellular elements within the OC TME, detailing their main functions and possible signaling pathways. Additionally, we examine immunotherapy rechallenge in OC, with a specific emphasis on the biological reasons behind resistance to ICIs.
Collapse
Affiliation(s)
- Jinxin Chen
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Lu Yang
- Department of Internal Medicine, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Yiming Ma
- Department of Medical Oncology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| | - Ye Zhang
- Department of Radiation Oncology, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| |
Collapse
|
24
|
Ahmed A, Joseph AM, Zhou J, Horn V, Uddin J, Lyu M, Goc J, Sockolow RE, Wing JB, Vivier E, Sakaguchi S, Sonnenberg GF. CTLA-4-expressing ILC3s restrain interleukin-23-mediated inflammation. Nature 2024; 630:976-983. [PMID: 38867048 PMCID: PMC11298788 DOI: 10.1038/s41586-024-07537-3] [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: 04/18/2023] [Accepted: 05/07/2024] [Indexed: 06/14/2024]
Abstract
Interleukin (IL-)23 is a major mediator and therapeutic target in chronic inflammatory diseases that also elicits tissue protection in the intestine at homeostasis or following acute infection1-4. However, the mechanisms that shape these beneficial versus pathological outcomes remain poorly understood. To address this gap in knowledge, we performed single-cell RNA sequencing on all IL-23 receptor-expressing cells in the intestine and their acute response to IL-23, revealing a dominance of T cells and group 3 innate lymphoid cells (ILC3s). Unexpectedly, we identified potent upregulation of the immunoregulatory checkpoint molecule cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) on ILC3s. This pathway was activated by gut microbes and IL-23 in a FOXO1- and STAT3-dependent manner. Mice lacking CTLA-4 on ILC3s exhibited reduced regulatory T cells, elevated inflammatory T cells and more-severe intestinal inflammation. IL-23 induction of CTLA-4+ ILC3s was necessary and sufficient to reduce co-stimulatory molecules and increase PD-L1 bioavailability on intestinal myeloid cells. Finally, human ILC3s upregulated CTLA-4 in response to IL-23 or gut inflammation and correlated with immunoregulation in inflammatory bowel disease. These results reveal ILC3-intrinsic CTLA-4 as an essential checkpoint that restrains the pathological outcomes of IL-23, suggesting that disruption of these lymphocytes, which occurs in inflammatory bowel disease5-7, contributes to chronic inflammation.
Collapse
Affiliation(s)
- Anees Ahmed
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Ann M Joseph
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jordan Zhou
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Veronika Horn
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jazib Uddin
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Mengze Lyu
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jeremy Goc
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Robbyn E Sockolow
- Department of Pediatrics, Division of Gastroenterology, Hepatology, & Nutrition, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - James B Wing
- Laboratory of Experimental Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Japan
- Laboratory of Human Single Cell Immunology, WPI IFReC, Osaka University, Suita, Japan
- Human Single Cell Immunology Team, Center for Infectious Disease Education and Research, Osaka University, Suita, Japan
| | - Eric Vivier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
- APHM, Hôpital de la Timone, Marseille-Immunopôle, Marseille, France
- Paris Saclay Cancer Cluster, Villejuif, France
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Japan
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Gregory F Sonnenberg
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
25
|
Liu Q, Pickett T, Hodge D, Rios C, Arnold M, Dong G, Hamilton SE, Rehermann B. Leveraging dirty mice that have microbial exposure to improve preclinical models of human immune status and disease. Nat Immunol 2024; 25:947-950. [PMID: 38750319 PMCID: PMC11656454 DOI: 10.1038/s41590-024-01842-9] [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] [Indexed: 06/05/2024]
Abstract
The National Institute of Allergy and Infectious Diseases (NIAID) hosted a two-day virtual workshop on leveraging microbial exposure to improve mouse models of human immune status and disease. The workshop’s objective was to evaluate the current state of knowledge in the field and to identify gaps, challenges and future directions.
Collapse
Affiliation(s)
- Qian Liu
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS, Rockville, MD, USA.
| | - Thames Pickett
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS, Rockville, MD, USA
| | - Deborah Hodge
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS, Rockville, MD, USA
| | - Carmen Rios
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS, Rockville, MD, USA
| | - Michelle Arnold
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS, Rockville, MD, USA
| | - Gang Dong
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS, Rockville, MD, USA
| | - Sara E Hamilton
- Department of Laboratory Medicine & Pathology, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Barbara Rehermann
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, USA.
| |
Collapse
|
26
|
Chen J, Wang D, Zhang H. Fc-null anti-CTLA-4 antibody: a novel strategy to facilitate cancer immunotherapy by ridding the colitis-inducing mishap. MedComm (Beijing) 2024; 5:e622. [PMID: 38881673 PMCID: PMC11176731 DOI: 10.1002/mco2.622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
Writing recently in Science, Lo and coworkers characterized a critical role of the gut microbiota in CTLA-4 blockade-induced colitis, revealing that an Fc domain deficient anti-CTLA-4 antibody can elicit antitumor responses effectively while avoiding the induction of colitis-like disease.1 This research opens up novel avenues for employing anti-CTLA-4 antibody therapy to circumvent the onset of colitis, which is often considered the Achilles' heel of what is arguably the most efficacious treatment for certain blood cancers and/or solid tumors.
Collapse
Affiliation(s)
- Jingjing Chen
- Department of Gastroenterology West China Hospital Sichuan University Chengdu China
- Department of Pediatrics The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou China
| | - Dexuan Wang
- Department of Pediatrics The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou China
| | - Hu Zhang
- Department of Gastroenterology West China Hospital Sichuan University Chengdu China
| |
Collapse
|
27
|
Jia D, Wang L. Modulating commensal bacteria to intervene in cancer immunotherapy. Clin Transl Med 2024; 14:e1704. [PMID: 38778442 PMCID: PMC11112531 DOI: 10.1002/ctm2.1704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Affiliation(s)
- Dingjiacheng Jia
- Department of GastroenterologySecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institution of GastroenterologyZhejiang UniversityHangzhouChina
| | - Liangjing Wang
- Department of GastroenterologySecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institution of GastroenterologyZhejiang UniversityHangzhouChina
- Cancer CenterZhejiang UniversityHangzhouChina
| |
Collapse
|
28
|
Ray K. A role for the gut microbiota in immune checkpoint inhibitor-induced colitis. Nat Rev Gastroenterol Hepatol 2024; 21:141. [PMID: 38263514 DOI: 10.1038/s41575-024-00897-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Affiliation(s)
- Katrina Ray
- Nature Reviews Gastroenterology & Hepatology, .
| |
Collapse
|
29
|
Apaza CJ, Días M, García Tejedor A, Boscá L, Laparra Llopis JM. Contribution of Nucleotide-Binding Oligomerization Domain-like (NOD) Receptors to the Immune and Metabolic Health. Biomedicines 2024; 12:341. [PMID: 38397943 PMCID: PMC10886542 DOI: 10.3390/biomedicines12020341] [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: 01/09/2024] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Nucleotide-binding oligomerization domain-like (NOD) receptors rely on the interface between immunity and metabolism. Dietary factors constitute critical players in the activation of innate immunity and modulation of the gut microbiota. The latter have been involved in worsening or improving the control and promotion of diseases such as obesity, type 2 diabetes, metabolic syndrome, diseases known as non-communicable metabolic diseases (NCDs), and the risk of developing cancer. Intracellular NODs play key coordinated actions with innate immune 'Toll-like' receptors leading to a diverse array of gene expressions that initiate inflammatory and immune responses. There has been an improvement in the understanding of the molecular and genetic implications of these receptors in, among others, such aspects as resting energy expenditure, insulin resistance, and cell proliferation. Genetic factors and polymorphisms of the receptors are determinants of the risk and severity of NCDs and cancer, and it is conceivable that dietary factors may have significant differential consequences depending on them. Host factors are difficult to influence, while environmental factors are predominant and approachable with a preventive and/or therapeutic intention in obesity, T2D, and cancer. However, beyond the recognition of the activation of NODs by peptidoglycan as its prototypical agonist, the underlying molecular response(s) and its consequences on these diseases remain ill-defined. Metabolic (re)programming is a hallmark of NCDs and cancer in which nutritional strategies might play a key role in preventing the unprecedented expansion of these diseases. A better understanding of the participation and effects of immunonutritional dietary ingredients can boost integrative knowledge fostering interdisciplinary science between nutritional precision and personalized medicine against cancer. This review summarizes the current evidence concerning the relationship(s) and consequences of NODs on immune and metabolic health.
Collapse
Affiliation(s)
- César Jeri Apaza
- Molecular Immunonutrition Group, Madrid Institute for Advanced Studies in Food (IMDEA Food), Ctra Cantoblanco, 8, 28049 Madrid, Spain;
| | - Marisol Días
- Center of Biological Enginneering (CEB), Iberian Nantotechnology Laboratory (INL), University of Minho, 4715-330 Braga, Portugal;
| | - Aurora García Tejedor
- Bioactivity and Nutritional Immunology Group (BIOINUT), Faculty of Health Sciences, Universidad Internacional de Valencia (VIU), Pintor Sorolla 21, 46002 Valencia, Spain;
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols-Morreale (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain;
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Melchor Fernández Almagro 6, 28029 Madrid, Spain
| | - José Moisés Laparra Llopis
- Molecular Immunonutrition Group, Madrid Institute for Advanced Studies in Food (IMDEA Food), Ctra Cantoblanco, 8, 28049 Madrid, Spain;
| |
Collapse
|