1
|
Shang J, Del Valle DM, Britton GJ, Mead K, Rajpal U, Chen-Liaw A, Mogno I, Li Z, Menon R, Gonzalez-Kozlova E, Elkrief A, Peled JU, Gonsalves TR, Shah NJ, Postow M, Colombel JF, Gnjatic S, Faleck DM, Faith JJ. Baseline colitogenicity and acute perturbations of gut microbiota in immunotherapy-related colitis. J Exp Med 2025; 222:e20232079. [PMID: 39666007 PMCID: PMC11636624 DOI: 10.1084/jem.20232079] [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: 11/13/2023] [Revised: 09/17/2024] [Accepted: 11/21/2024] [Indexed: 12/13/2024] Open
Abstract
Immunotherapy-related colitis (irC) frequently emerges as an immune-related adverse event during immune checkpoint inhibitor therapy and is presumably influenced by the gut microbiota. We longitudinally studied microbiomes from 38 ICI-treated cancer patients. We compared 13 ICI-treated subjects who developed irC against 25 ICI-treated subjects who remained irC-free, along with a validation cohort. Leveraging a preclinical mouse model, predisease stools from irC subjects induced greater colitigenicity upon transfer to mice. The microbiota during the first 10 days of irC closely resembled inflammatory bowel disease microbiomes, with reduced diversity, increased Proteobacteria and Veillonella, and decreased Faecalibacterium, which normalized before irC remission. These findings highlight the irC gut microbiota as functionally distinct but phylogenetically similar to non-irC and healthy microbiomes, with the exception of an acute, transient disruption early in irC. We underscore the significance of longitudinal microbiome profiling in developing clinical avenues to detect, monitor, and mitigate irC in ICI therapy cancer patients.
Collapse
Affiliation(s)
- Joan Shang
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Diane Marie Del Valle
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Graham J. Britton
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - K.R. Mead
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Urvija Rajpal
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alice Chen-Liaw
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ilaria Mogno
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhihua Li
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Edgar Gonzalez-Kozlova
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Arielle Elkrief
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan U. Peled
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Tina Ruth Gonsalves
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Neil J. Shah
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Michael Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Jean-Frederic Colombel
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sacha Gnjatic
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David M. Faleck
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Jeremiah J. Faith
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
2
|
Hanahan D, Michielin O, Pittet MJ. Convergent inducers and effectors of T cell paralysis in the tumour microenvironment. Nat Rev Cancer 2025; 25:41-58. [PMID: 39448877 DOI: 10.1038/s41568-024-00761-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/23/2024] [Indexed: 10/26/2024]
Abstract
Tumorigenesis embodies the formation of a heterotypic tumour microenvironment (TME) that, among its many functions, enables the evasion of T cell-mediated immune responses. Remarkably, most TME cell types, including cancer cells, fibroblasts, myeloid cells, vascular endothelial cells and pericytes, can be stimulated to deploy immunoregulatory programmes. These programmes involve regulatory inducers (signals-in) and functional effectors (signals-out) that impair CD8+ and CD4+ T cell activity through cytokines, growth factors, immune checkpoints and metabolites. Some signals target specific cell types, whereas others, such as transforming growth factor-β (TGFβ) and prostaglandin E2 (PGE2), exert broad, pleiotropic effects; as signals-in, they trigger immunosuppressive programmes in most TME cell types, and as signals-out, they directly inhibit T cells and also modulate other cells to reinforce immunosuppression. This functional diversity and redundancy pose a challenge for therapeutic targeting of the immune-evasive TME. Fundamentally, the commonality of regulatory programmes aimed at abrogating T cell activity, along with paracrine signalling between cells of the TME, suggests that many normal cell types are hard-wired with latent functions that can be triggered to prevent inappropriate immune attack. This intrinsic capability is evidently co-opted throughout the TME, enabling tumours to evade immune destruction.
Collapse
Affiliation(s)
- Douglas Hanahan
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, Switzerland.
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.
| | - Olivier Michielin
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
- Department of Medicine, University of Geneva (UNIGE), Geneva, Switzerland
| | - Mikael J Pittet
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
- Department of Pathology and Immunology, University of Geneva (UNIGE), Geneva, Switzerland
| |
Collapse
|
3
|
Tian C, Yuan X, Li X, Li Z. Understanding the link between respiratory microbiota and asthma. Asian J Surg 2024; 47:5411-5413. [PMID: 38944607 DOI: 10.1016/j.asjsur.2024.06.079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/18/2024] [Indexed: 07/01/2024] Open
Affiliation(s)
- Chunyuan Tian
- Department of Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150040, China; Department of Respiratory Medicine, The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Xingxing Yuan
- Department of Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150040, China; Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, 150006, China.
| | - Xing Li
- Department of Respiratory Medicine, The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| | - Zhuying Li
- Department of Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150040, China; Department of Respiratory Medicine, The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| |
Collapse
|
4
|
Deng F, Ye H, Zhang P, Xu J, Li Y, Sun M, Yang Z. Association Between Antibiotic and Outcomes of Chemoimmunotherapy for Extensive-Stage Small Cell Lung Cancer: A Multicenter Retrospective Study of 132 Patients. Thorac Cancer 2024. [PMID: 39566958 DOI: 10.1111/1759-7714.15492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024] Open
Abstract
INTRODUCTION To evaluate the impact of antibiotic (ATB) exposure on the outcome of chemoimmunotherapy in patients with extensive-stage small cell lung cancer (ES-SCLC). METHODS In this multicenter retrospective study, 132 patients with ES-SCLC who received chemoimmunotherapy were included from three hospitals in China. Patients receiving ATB within 30 days prior to initiating ICI therapy (p-ATB) and those receiving concurrent ICI therapy until cessation (c-ATB)were compared to those who did not (n-ATB). Progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and immune-related adverse events (irAEs) were assessed. To avoid immortal time bias, c-ATB was analyzed as a time-dependent covariate in the Cox proportional hazards model. RESULTS Among the 132 patients, 25 were included in the p-ATB group and 26 in the c-ATB group, while 81 patients were categorized in the n-ATB group. Multivariate analysis revealed no significant differences in PFS (aHR = 1.028, 95% CI: 0.666-1.589, p = 0.900) and OS (aHR = 0.957, 95% CI: 0.549-1.668, p = 0.877) between the p-ATB and n-ATB groups. Similarly, p-ATB had no significant impact on ORR (p = 0.510) or irAEs (p = 0.516). The use of c-ATB had no significant effect on either PFS (aHR: 1.165, 95% CI: 0.907-1.497; p = 0.232) or OS (aHR: 1.221, 95% CI: 0.918-1.624; p = 0.171) by multivariate analysis. CONCLUSIONS p-ATB has no significant impact on PFS, OS, ORR, or the incidence of irAEs in ES-SCLC patients receiving chemoimmunotherapy. Similarly, c-ATB does not seem to affect PFS or OS.
Collapse
Affiliation(s)
- Fang Deng
- Department of Oncology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong, China
| | - Hong Ye
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong, China
| | - Ping Zhang
- Department of Oncology, Binzhou People's Hospital, Binzhou, Shandong, China
| | - Jing Xu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong, China
| | - Yu Li
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Meiling Sun
- Department of Respiratory Medicine, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, China
| | - Zhongfei Yang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong, China
| |
Collapse
|
5
|
Sevcikova A, Martiniakova M, Omelka R, Stevurkova V, Ciernikova S. The Link Between the Gut Microbiome and Bone Metastasis. Int J Mol Sci 2024; 25:12086. [PMID: 39596154 PMCID: PMC11593804 DOI: 10.3390/ijms252212086] [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: 10/30/2024] [Revised: 11/08/2024] [Accepted: 11/09/2024] [Indexed: 11/28/2024] Open
Abstract
The gut microbiome is essential for regulating host metabolism, defending against pathogens, and shaping the host's immune system. Mounting evidence highlights that disruption in gut microbial communities significantly impacts cancer development and treatment. Moreover, tumor-associated microbiota, along with its metabolites and toxins, may contribute to cancer progression by promoting epithelial-to-mesenchymal transition, angiogenesis, and metastatic spread to distant organs. Bones, in particular, are common sites for metastasis due to a rich supply of growth and neovascularization factors and extensive blood flow, especially affecting patients with thyroid, prostate, breast, lung, and kidney cancers, where bone metastases severely reduce the quality of life. While the involvement of the gut microbiome in bone metastasis formation is still being explored, proposed mechanisms suggest that intestinal dysbiosis may alter the bone microenvironment via the gut-immune-bone axis, fostering a premetastatic niche and immunosuppressive milieu suitable for cancer cell colonization. Disruption in the delicate balance of bone modeling and remodeling may further create a favorable environment for metastatic growth. This review focuses on the link between beneficial or dysbiotic microbiome composition and bone homeostasis, as well as the role of the microbiome in bone metastasis development. It also provides an overview of clinical trials evaluating the impact of gut microbial community structure on bone parameters across various conditions or health-related issues. Dietary interventions and microbiota modulation via probiotics, prebiotics, and fecal microbiota transplantation help support bone health and might offer promising strategies for addressing bone-related complications in cancer.
Collapse
Affiliation(s)
- Aneta Sevcikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (A.S.); (V.S.)
| | - Monika Martiniakova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia;
| | - Radoslav Omelka
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia;
| | - Viola Stevurkova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (A.S.); (V.S.)
| | - Sona Ciernikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (A.S.); (V.S.)
| |
Collapse
|
6
|
Liu T, Gu Y, Zhang Y, Li Y. Integrin α2 in the microenvironment and the tumor compartment of digestive (gastrointestinal) cancers: emerging regulators and therapeutic opportunities. Front Oncol 2024; 14:1439709. [PMID: 39568561 PMCID: PMC11576383 DOI: 10.3389/fonc.2024.1439709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024] Open
Abstract
Integrins are a family of cell surface membrane receptors and play a crucial role in facilitating bidirectional cell signaling. Integrin α2 (ITGA2) is expressed across a range of cell types, including epithelial cells, platelets, megakaryocytes, and fibroblasts, where it functions as a surface marker and it is implicated in the cell movements. The most recent findings have indicated that ITAG2 has the potential to function as a novel regulatory factor in cancer, responsible for driving tumorigenesis, inducing chemoresistance, regulating genomic instability and remodeling tumor microenvironment. Hence, we primarily focus on elucidating the biological function and mechanism of ITGA2 within the digestive tumor microenvironment, while highlighting its prospective utilization as a therapeutic target for cancer therapy.
Collapse
Affiliation(s)
- Tiantian Liu
- Lanzhou University Second Hospital, the Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Yanmei Gu
- Lanzhou University Second Hospital, the Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Yuyu Zhang
- Lanzhou University Second Hospital, the Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Yumin Li
- Lanzhou University Second Hospital, the Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
7
|
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
|
8
|
Bredon M, Le Malicot K, Louvet C, Evesque L, Gonzalez D, Tougeron D, Sokol H. Faecalibacteriumprausnitzii Is Associated With Clinical Response to Immune Checkpoint Inhibitors in Patients With Advanced Gastric Adenocarcinoma: Results of Microbiota Analysis of PRODIGE 59-FFCD 1707-DURIGAST Trial. Gastroenterology 2024:S0016-5085(24)05594-X. [PMID: 39454892 DOI: 10.1053/j.gastro.2024.10.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024]
Affiliation(s)
- Marius Bredon
- Sorbonne Université, Institut National de la Santé et de la Recherche Médical, Gastroenterology Department, Centre de Recherche Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, Paris, France; Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, France
| | - Karine Le Malicot
- Fédération Francophone de Cancérologie Digestive, EPICAD, Institut National de la Santé et de la Recherche Médical, LNC-UMR 1231, Bourgogne Franche-Comté University, Dijon, France
| | - Christophe Louvet
- Department of Medical Oncology, Institute Mutualiste Montsouris, Paris, France
| | - Ludovic Evesque
- Department of Digestive Oncology, Antoine Lacassagne Centre, Nice, France
| | - Daniel Gonzalez
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, France
| | - David Tougeron
- Department of Gastroenterology and Hepatology, Poitiers University Hospital, Poitiers, France.
| | - Harry Sokol
- Sorbonne Université, Institut National de la Santé et de la Recherche Médical, Gastroenterology Department, Centre de Recherche Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, Paris, France; Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, France; Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France.
| |
Collapse
|
9
|
Lee WS, Lee SJ, Lee HJ, Yang H, Go EJ, Gansukh E, Song KH, Xiang X, Park DG, Alain T, Chon HJ, Kim C. Oral reovirus reshapes the gut microbiome and enhances antitumor immunity in colon cancer. Nat Commun 2024; 15:9092. [PMID: 39438458 PMCID: PMC11496807 DOI: 10.1038/s41467-024-53347-6] [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: 09/19/2023] [Accepted: 10/02/2024] [Indexed: 10/25/2024] Open
Abstract
The route of oncolytic virotherapy is pivotal for immunotherapeutic efficacy in advanced cancers. In this preclinical study, an oncolytic reovirus (RC402) is orally administered to induce antitumor immunity. Oral reovirus treatment shows no gross toxicities and effectively suppresses multifocal tumor lesions. Orally administered reovirus interacts with the host immune system in the Peyer's patch of the terminal ileum, increases IgA+ antibody-secreting cells in the lamina propria through MAdCAM-1+ blood vessels, and reshapes the gut microbiome. Oral reovirus promotes antigen presentation, type I/II interferons, and T cell activation within distant tumors, but does not reach or directly infect tumor cells beyond the gastrointestinal tract. In contrast to intratumoral reovirus injection, the presence of the gut microbiome, Batf3+ dendritic cells, type I interferons, and CD8+ T cells are indispensable for orally administered reovirus-induced antitumor immunity. Oral reovirus treatment is most effective when combined with αPD-1(L1) and/or αCTLA-4, leading to complete colon tumor regression and protective immune memory. Collectively, oral reovirus virotherapy is a feasible and effective immunotherapeutic strategy in preclinical studies.
Collapse
Affiliation(s)
- Won Suk Lee
- Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-do, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University, Seongnam, Gyeonggi-do, Republic of Korea
| | - Seung Joon Lee
- Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-do, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University, Seongnam, Gyeonggi-do, Republic of Korea
| | - Hye Jin Lee
- Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-do, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University, Seongnam, Gyeonggi-do, Republic of Korea
| | - Hannah Yang
- Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-do, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University, Seongnam, Gyeonggi-do, Republic of Korea
| | - Eun-Jin Go
- Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-do, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University, Seongnam, Gyeonggi-do, Republic of Korea
| | | | | | - Xiao Xiang
- Department of Biochemistry, Microbiology, and Immunology, Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Dong Guk Park
- Virocure Inc., Seoul, Republic of Korea
- Department of Surgery, School of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Tommy Alain
- Department of Biochemistry, Microbiology, and Immunology, Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Hong Jae Chon
- Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-do, Republic of Korea.
- Laboratory of Translational Immuno-Oncology, CHA University, Seongnam, Gyeonggi-do, Republic of Korea.
| | - Chan Kim
- Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-do, Republic of Korea.
- Laboratory of Translational Immuno-Oncology, CHA University, Seongnam, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
10
|
Praveen Z, Choi SW, Lee JH, Park JY, Oh HJ, Kwon IJ, Park JH, Kim MK. Oral Microbiome and CPT1A Function in Fatty Acid Metabolism in Oral Cancer. Int J Mol Sci 2024; 25:10890. [PMID: 39456670 PMCID: PMC11508181 DOI: 10.3390/ijms252010890] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/26/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
The oral microbiome is crucial for human health. Although oral dysbiosis may contribute to oral cancer (OC), the detailed relationships between the microbiome and OC remain unclear. In this case-control study, we aimed to elucidate the connection between the oral microbiome and mechanisms potentially involved in oral cancer. The study analyzed 1022 oral saliva samples, including 157 from oral cancer patients and 865 from healthy controls, using 16S ribosomal RNA (16S rRNA) sequencing and a Light Gradient Boosting Machine (LightGBM) model to identify four bacterial genera significantly associated with oral cancer. In patients with oral cancer, the relative abundance of Streptococcus and Parvimonas was higher; Corynebacterium and Prevotella showed decreased relative abundance; and levels of fatty acid oxidation enzymes, including Carnitine palmitoyltransferase 1A (CPT1A), long-chain acyl-CoA synthetase, acyl-CoA dehydrogenase, diacylglycerol choline phosphotransferase, and H+-transporting ATPase, were significantly higher compared to controls. Conversely, healthy controls exhibited increased levels of short-chain fatty acids (SCFAs) and CD4+T-helper cell counts. Survival analysis revealed that higher abundance of Streptococcus and Parvimonas, which correlated positively with interleukin-6, tumor necrosis factor-alpha, and CPT1A, were linked to poorer disease-free survival (DFS) and overall survival (OS) rates, while Prevotella and Corynebacterium were associated with better outcomes. These findings suggest that changes in these bacterial genera are associated with alterations in specific cytokines, CPT1A levels, SCFAs in oral cancer, with lower SCFA levels in patients reinforcing this link. Overall, these microbiome changes, along with cytokine and enzyme alterations, may serve as predictive markers, enhancing diagnostic accuracy for oral cancer.
Collapse
Affiliation(s)
- Zeba Praveen
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Republic of Korea;
| | - Sung-Weon Choi
- Oral Oncology Clinic, Research Institute and Hospital, National Cancer Center, 323 Ilsandong-gu, Goyang-si 10408, Gyeonggi-do, Republic of Korea; (S.-W.C.); (J.H.L.); (J.Y.P.); (H.J.O.)
| | - Jong Ho Lee
- Oral Oncology Clinic, Research Institute and Hospital, National Cancer Center, 323 Ilsandong-gu, Goyang-si 10408, Gyeonggi-do, Republic of Korea; (S.-W.C.); (J.H.L.); (J.Y.P.); (H.J.O.)
| | - Joo Yong Park
- Oral Oncology Clinic, Research Institute and Hospital, National Cancer Center, 323 Ilsandong-gu, Goyang-si 10408, Gyeonggi-do, Republic of Korea; (S.-W.C.); (J.H.L.); (J.Y.P.); (H.J.O.)
| | - Hyun Jun Oh
- Oral Oncology Clinic, Research Institute and Hospital, National Cancer Center, 323 Ilsandong-gu, Goyang-si 10408, Gyeonggi-do, Republic of Korea; (S.-W.C.); (J.H.L.); (J.Y.P.); (H.J.O.)
| | - Ik Jae Kwon
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea;
| | - Jin Hee Park
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Republic of Korea;
| | - Mi Kyung Kim
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Republic of Korea;
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
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] [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
|
14
|
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
|
15
|
Zhao M, Tian J, Hou W, Yin L, Li W. Global research trends on the associations between the microbiota and lung cancer: a visualization bibliometric analysis (2008-2023). Front Microbiol 2024; 15:1416385. [PMID: 39282557 PMCID: PMC11392740 DOI: 10.3389/fmicb.2024.1416385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Numerous papers have been published on the microbiota in lung cancer in recent years. However, there is still a lack of bibliometric analysis of the microbiota in lung cancer in this field. Our paper did bibliometric analyses and elucidated the knowledge structure and study hotspots related to the microbiota in lung cancer patients. We screened publications reporting on the microbiota in lung cancer from 2008 to 2023 from the Web of Science Core Collection (WoSCC) database, and carried out bibliometric analyses by the application of the VOSviewers, CiteSpace and R package "bibliometrix." The 684 documents enrolled in the analysis were obtained from 331 institutions in 67 regions by 4,661 authors and were recorded in 340 journals. Annual papers are growing rapidly, and the countries of China, the United States and Italy are contributing the most to this area of research. Zhejiang University is the main research organization. Science and Cancer had significant impacts on this area. Zhang Yan had the most articles, and the Bertrand Routy had the most co-cited times. Exploring the mechanism of action of the lung and/or gut microbiota in lung cancer and therapeutic strategies involving immune checkpoint inhibitors in lung cancer are the main topics. Moreover, "gut microbiota," "immunotherapy," and "short-chain fatty acids" are important keywords for upcoming study hotspots. In conclusion, microbiota research offers promising opportunities in lung cancer, with pivotal studies exploring the mechanisms that link lung and gut microbiota to therapeutic strategies, particularly through immune checkpoint inhibitors. Moreover, the gut-lung axis emerges as a novel target for innovative treatments. Further research is essential to unravel the detailed mechanisms of this connection.
Collapse
Affiliation(s)
- Maoyuan Zhao
- Lung Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jie Tian
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wang Hou
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Liyuan Yin
- Lung Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, China
| |
Collapse
|
16
|
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
|
17
|
Shakhpazyan NK, Mikhaleva LM, Bedzhanyan AL, Gioeva ZV, Mikhalev AI, Midiber KY, Pechnikova VV, Biryukov AE. Exploring the Role of the Gut Microbiota in Modulating Colorectal Cancer Immunity. Cells 2024; 13:1437. [PMID: 39273009 PMCID: PMC11394638 DOI: 10.3390/cells13171437] [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: 07/26/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
The gut microbiota plays an essential role in maintaining immune homeostasis and influencing the immune landscape within the tumor microenvironment. This review aims to elucidate the interactions between gut microbiota and tumor immune dynamics, with a focus on colorectal cancer (CRC). The review spans foundational concepts of immuno-microbial interplay, factors influencing microbiome composition, and evidence linking gut microbiota to cancer immunotherapy outcomes. Gut microbiota modulates anti-cancer immunity through several mechanisms, including enhancement of immune surveillance and modulation of inflammatory responses. Specific microbial species and their metabolic byproducts can significantly influence the efficacy of cancer immunotherapies. Furthermore, microbial diversity within the gut microbiota correlates with clinical outcomes in CRC, suggesting potential as a valuable biomarker for predicting response to immunotherapy. Conclusions: Understanding the relationship between gut microbiota and tumor immune responses offers potential for novel therapeutic strategies and biomarker development. The gut microbiota not only influences the natural history and treatment response of CRC but also serves as a critical modulator of immune homeostasis and anti-cancer activity. Further exploration into the microbiome's role could enhance the effectiveness of existing treatments and guide the development of new therapeutic modalities.
Collapse
Affiliation(s)
- Nikolay K Shakhpazyan
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Center of Surgery, 119435 Moscow, Russia
| | - Liudmila M Mikhaleva
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Center of Surgery, 119435 Moscow, Russia
| | - Arkady L Bedzhanyan
- Department of Abdominal Surgery and Oncology II (Coloproctology and Uro-Gynecology), Petrovsky National Research Center of Surgery, 119435 Moscow, Russia
| | - Zarina V Gioeva
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Center of Surgery, 119435 Moscow, Russia
| | - Alexander I Mikhalev
- Department of Hospital Surgery No. 2, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Konstantin Y Midiber
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Center of Surgery, 119435 Moscow, Russia
- Institute of Medicine, Peoples' Friendship University of Russia named after Patrice Lumumba, 6 Miklukho-Maklaya St., 117198 Moscow, Russia
| | - Valentina V Pechnikova
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Center of Surgery, 119435 Moscow, Russia
| | - Andrey E Biryukov
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Center of Surgery, 119435 Moscow, Russia
| |
Collapse
|
18
|
Chen Y, Che X, Rong Y, Zhu J, Yu Y, Xu H, Sun Y, Chen H, Yan L, Chen L, Xu Y, Zhang J. Immunomodulation in Endometriosis: Investigating the interrelationship between VISTA expression and Escherichia.Shigella-Associated metabolites. Int Immunopharmacol 2024; 137:112366. [PMID: 38852526 DOI: 10.1016/j.intimp.2024.112366] [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: 04/02/2024] [Revised: 05/18/2024] [Accepted: 05/27/2024] [Indexed: 06/11/2024]
Abstract
AIMS Endometriosis is characterized by an abnormal immune microenvironment. Despite the extensive use of immune therapies, the application of immune checkpoint inhibitors in endometriosis lacks confidence due to the instability of preclinical research data. This study aims to elucidate the regulation of the immune inhibitory checkpoint VISTA and its effects on T cells from the perspective of microbiota and metabolism. MAIN METHODS We divided endometriosis patients into high and low groups based on the expression levels of VISTA in lesion tissues. We collected peritoneal fluid samples from these two groups and performed 16 s RNA sequencing and metabolomics analysis to investigate microbial diversity and differential metabolites. Through combined analysis, we identified microbial-associated metabolites and validated their correlation with VISTA and CD8 + T cells using ELISA and immunofluorescence. In vitro experiments were conducted to confirm the regulatory relationship among these factors. KEY FINDINGS Our findings revealed a distinct correlation between VISTA expression and the microbial colony Escherichia.Shigella. Moreover, we identified the metabolites LTD4-d5 and 2-n-Propylthiazolidine-4-carboxylic acid as being associated with both Escherichia.Shigella and VISTA expression. In vitro experiments confirmed the inhibitory effects of these metabolites on VISTA expression, while they demonstrated a positive regulation of CD8 + T cell infiltration into endometriotic lesions. SIGNIFICANCE This study reveals the connection between microbial diversity, metabolites, and VISTA expression in the immune microenvironment of endometriosis, providing potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Yichen Chen
- Women and Children's Hospital of Ningbo University, Ningbo, China
| | - Xuan Che
- Jiaxing University Affiliated Women and Children Hospital, Jiaxing, China
| | - Yishen Rong
- Women and Children's Hospital of Ningbo University, Ningbo, China; Ningbo University, Ningbo, China
| | - Jue Zhu
- Women and Children's Hospital of Ningbo University, Ningbo, China
| | - Yayuan Yu
- Jiaxing University Affiliated Women and Children Hospital, Jiaxing, China
| | - Hong Xu
- International Peace Maternity and Child Health Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, China
| | - Yuhui Sun
- Women and Children's Hospital of Ningbo University, Ningbo, China
| | - Huan Chen
- Women and Children's Hospital of Ningbo University, Ningbo, China; Ningbo University, Ningbo, China
| | - Lifeng Yan
- Women and Children's Hospital of Ningbo University, Ningbo, China
| | - Liang Chen
- Women and Children's Hospital of Ningbo University, Ningbo, China
| | - Yanan Xu
- Women and Children's Hospital of Ningbo University, Ningbo, China
| | - Jing Zhang
- Women and Children's Hospital of Ningbo University, Ningbo, China.
| |
Collapse
|
19
|
Golshani M, Taylor JA, Woolbright BL. Understanding the microbiome as a mediator of bladder cancer progression and therapeutic response. Urol Oncol 2024:S1078-1439(24)00541-6. [PMID: 39117491 DOI: 10.1016/j.urolonc.2024.07.004] [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: 04/02/2024] [Revised: 05/17/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024]
Abstract
Bladder cancer (BCa) remains a significant source of morbidity and mortality. BCa is one of the most expensive tumors to treat, in part because of a lack of nonsurgical options. The recent advent of immunotherapy, alone or in combination with other compounds, has improved therapeutic options. Resistance to immunotherapy remains common, and many patients do not have durable response. Recent advances indicate immunotherapy efficacy may be tied in part to the endogenous bacteria present in our body, more commonly referred to as the microbiome. Laboratory and clinical data now support the idea that a healthy microbiome is critical to effective response to immunotherapy. At the same time, pathogenic interactions between the microbiome and immune cells can also serve to drive formation of tumors, increasing the complexity of these interactions. Given the rising importance of immunotherapy in BCa, understanding how we might be able to alter the microbiome to improve therapeutic efficacy offers a novel route to improved patient care. The goal of this review is to examine our current understanding of microbial interactions with the immune system and cancer with an emphasis on BCa. We will further attempt to define both current gaps in knowledge and future directions that may yield beneficial results to the field.
Collapse
Affiliation(s)
- Mahgol Golshani
- School of Medicine, University of Kansas Medical Center, Kansas City, KS
| | - John A Taylor
- Department of Urology, University of Kansas Medical Center, Kansas City, KS; Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS
| | | |
Collapse
|
20
|
Wu B, Quan C, He Y, Matsika J, Huang J, Liu B, Chen J. Targeting gut and intratumoral microbiota: a novel strategy to improve therapy resistance in cancer with a focus on urologic tumors. Expert Opin Biol Ther 2024; 24:747-759. [PMID: 38910461 DOI: 10.1080/14712598.2024.2371543] [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: 04/07/2024] [Accepted: 06/19/2024] [Indexed: 06/25/2024]
Abstract
INTRODUCTION Growing attention has been drawn to urologic tumors due to their rising incidence and suboptimal clinical treatment outcomes. Cancer therapy resistance poses a significant challenge in clinical oncology, limiting the efficacy of conventional treatments and contributing to disease progression. Recent research has unveiled a complex interplay between the host microbiota and cancer cells, highlighting the role of the microbiota in modulating therapeutic responses. AREAS COVERED We used the PubMed and Web of Science search engines to identify key publications in the fields of tumor progression and urologic tumor treatment, specifically focusing on the role of the microbiota. In this review, we summarize the current literature on how microbiota influence the tumor microenvironment and anti-tumor immunity, as well as their impact on treatments for urinary system malignancies, highlighting promising future applications. EXPERT OPINION We explore how the composition and function of the gut microbiota influence the tumor microenvironment and immune response, ultimately impacting treatment outcomes. Additionally, we discuss emerging strategies targeting the microbiota to enhance therapeutic efficacy and overcome resistance. The application of antibiotics, fecal microbiota transplantation, and oncolytic bacteria has improved tumor treatment outcomes, which provides a novel insight into developing therapeutic strategies for urologic cancer.
Collapse
Affiliation(s)
- Bingquan Wu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chao Quan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yunbo He
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juliet Matsika
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jinliang Huang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bolong Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jinbo Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
21
|
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
|
22
|
Kyriazi AA, Karaglani M, Agelaki S, Baritaki S. Intratumoral Microbiome: Foe or Friend in Reshaping the Tumor Microenvironment Landscape? Cells 2024; 13:1279. [PMID: 39120310 PMCID: PMC11312414 DOI: 10.3390/cells13151279] [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: 06/05/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
The role of the microbiome in cancer and its crosstalk with the tumor microenvironment (TME) has been extensively studied and characterized. An emerging field in the cancer microbiome research is the concept of the intratumoral microbiome, which refers to the microbiome residing within the tumor. This microbiome primarily originates from the local microbiome of the tumor-bearing tissue or from translocating microbiome from distant sites, such as the gut. Despite the increasing number of studies on intratumoral microbiome, it remains unclear whether it is a driver or a bystander of oncogenesis and tumor progression. This review aims to elucidate the intricate role of the intratumoral microbiome in tumor development by exploring its effects on reshaping the multileveled ecosystem in which tumors thrive, the TME. To dissect the complexity and the multitude of layers within the TME, we distinguish six specialized tumor microenvironments, namely, the immune, metabolic, hypoxic, acidic, mechanical and innervated microenvironments. Accordingly, we attempt to decipher the effects of the intratumoral microbiome on each specialized microenvironment and ultimately decode its tumor-promoting or tumor-suppressive impact. Additionally, we portray the intratumoral microbiome as an orchestrator in the tumor milieu, fine-tuning the responses in distinct, specialized microenvironments and remodeling the TME in a multileveled and multifaceted manner.
Collapse
Affiliation(s)
- Athina A. Kyriazi
- Laboratory of Experimental Oncology, Division of Surgery, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Makrina Karaglani
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
- Laboratory of Hygiene and Environmental Protection, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Sofia Agelaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Stavroula Baritaki
- Laboratory of Experimental Oncology, Division of Surgery, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| |
Collapse
|
23
|
Koleva P, He J, Dunsmore G, Bozorgmehr N, Lu J, Huynh M, Tollenaar S, Huang V, Walter J, Way SS, Elahi S. CD71 + erythroid cells promote intestinal symbiotic microbial communities in pregnancy and neonatal period. MICROBIOME 2024; 12:142. [PMID: 39080725 PMCID: PMC11290123 DOI: 10.1186/s40168-024-01859-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 06/15/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND The establishment of microbial communities in neonatal mammals plays a pivotal role in shaping their immune responses to infections and other immune-related conditions. This process is influenced by a combination of endogenous and exogenous factors. Previously, we reported that depletion of CD71 + erythroid cells (CECs) results in an inflammatory response to microbial communities in newborn mice. RESULTS Here, we systemically tested this hypothesis and observed that the small intestinal lamina propria of neonatal mice had the highest frequency of CECs during the early days of life. This high abundance of CECs was attributed to erythropoiesis niches within the small intestinal tissues. Notably, the removal of CECs from the intestinal tissues by the anti-CD71 antibody disrupted immune homeostasis. This disruption was evident by alteration in the expression of antimicrobial peptides (AMPs), toll-like receptors (TLRs), inflammatory cytokines/chemokines, and resulting in microbial dysbiosis. Intriguingly, these alterations in microbial communities persisted when tested 5 weeks post-treatment, with a more notable effect observed in female mice. This illustrates a sex-dependent association between CECs and neonatal microbiome modulation. Moreover, we extended our studies on pregnant mice, observing that modulating CECs substantially alters the frequency and diversity of their microbial communities. Finally, we found a significantly lower proportion of CECs in the cord blood of pre-term human newborns, suggesting a potential role in dysregulated immune responses to microbial communities in the gut. CONCLUSIONS Our findings provide novel insights into pivotal role of CECs in immune homeostasis and swift adaptation of microbial communities in newborns. Despite the complexity of the cellular biology of the gut, our findings shed light on the previously unappreciated role of CECs in the dialogue between the microbiota and immune system. These findings have significant implications for human health. Video Abstract.
Collapse
Affiliation(s)
- Petya Koleva
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, Edmonton, Canada
| | - Jia He
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, Edmonton, Canada
| | - Garett Dunsmore
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, Edmonton, Canada
| | - Najmeh Bozorgmehr
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, Edmonton, Canada
| | - Julia Lu
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, Edmonton, Canada
| | - Maia Huynh
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, Edmonton, Canada
| | - Stephanie Tollenaar
- Department of Agricultural, Food & Nutritional Sciences, Edmonton, University of Alberta, Edmonton, Canada
| | - Vivian Huang
- Division of Gastroenterology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Division of Gastroenterology, Mount Sinai Hospital, Toronto, Canada
| | - Jens Walter
- Department of Agricultural, Food & Nutritional Sciences, Edmonton, University of Alberta, Edmonton, Canada
- School of Microbiology and Department of Medicine, APC Microbiome Ireland, University College Cork, National University of Ireland, Cork, Ireland
| | - Sing Sing Way
- Centre for Inflammation and Tolerance, Cincinnati Childrens Hospital, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Shokrollah Elahi
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, Edmonton, Canada.
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada.
- Glycomics Institute of Alberta, University of Alberta, Edmonton, Canada.
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.
- Alberta Transplant Institute, Edmonton, AB, Canada.
- 7020G Katz Group Centre for Pharmacology and Health Research, 11361-87Th Ave NW, Edmonton, AB, T6G2E1, Canada.
| |
Collapse
|
24
|
Elkrief A, Méndez-Salazar EO, Maillou J, Vanderbilt CM, Gogia P, Desilets A, Messaoudene M, Kelly D, Ladanyi M, Hellmann MD, Zitvogel L, Rudin CM, Routy B, Derosa L, Schoenfeld AJ. Antibiotics are associated with worse outcomes in lung cancer patients treated with chemotherapy and immunotherapy. NPJ Precis Oncol 2024; 8:143. [PMID: 39014160 PMCID: PMC11252311 DOI: 10.1038/s41698-024-00630-w] [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: 09/20/2023] [Accepted: 06/13/2024] [Indexed: 07/18/2024] Open
Abstract
Anti-PD(L)-1 inhibition combined with platinum doublet chemotherapy (Chemo-IO) has become the most frequently used standard of care regimen in patients with non-small cell lung cancer (NSCLC). The negative impact of antibiotics on clinical outcomes prior to anti-PD(L)-1 inhibition monotherapy (IO) has been demonstrated in multiple studies, but the impact of antibiotic exposure prior to initiation of Chemo-IO is controversial. We assessed antibiotic exposures at two time windows: within 60 days prior to therapy (-60 d window) and within 60 days prior to therapy and 42 days after therapy (-60 + 42d window) in 2028 patients with advanced NSCLC treated with Chemo-IO and IO monotherapy focusing on objective response rate (ORR: rate of partial response and complete response), progression-free survival (PFS), and overall survival (OS). We also assessed impact of antibiotic exposure in an independent cohort of 53 patients. Univariable and multivariable analyses were conducted along with a meta-analysis from similar studies. For the -60 d window, in the Chemo-IO group (N = 769), 183 (24%) patients received antibiotics. Antibiotic exposure was associated with worse ORR (27% vs 40%, p = 0.001), shorter PFS (3.9 months vs. 5.9 months, hazard ratio [HR] 1.35, 95%CI 1.1,1.6, p = 0.0012), as well as shorter OS (10 months vs. 15 months, HR 1.50, 95%CI 1.2,1.8, p = 0.00014). After adjusting for known prognostic factors in NSCLC, antibiotic exposure was independently associated with worse PFS (HR 1.39, 95%CI 1.35,1.7, p = 0.002) and OS (HR 1.61, 95%CI 1.28,2.03, p < 0.001). Similar results were obtained in the -60 + 42d window, and also in an independent cohort. In a meta-analysis of patients with NSCLC treated with Chemo-IO (N = 4) or IO monotherapy (N = 13 studies) antibiotic exposure before treatment was associated with worse OS among all patients (n = 11,351) (HR 1.93, 95% CI 1.52, 2.45) and Chemo-IO-treated patients (n = 1201) (HR 1.54, 95% CI 1.28, 1.84). Thus, antibiotics exposure prior to Chemo-IO is common and associated with worse outcomes, even after adjusting for other factors. These results highlight the need to implement antibiotic stewardship in routine oncology practice.
Collapse
Affiliation(s)
- Arielle Elkrief
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- University of Montreal Research Center (CR-CHUM), Montreal, QC, Canada.
- Department of Hematology-Oncology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada.
| | | | - Jade Maillou
- University of Montreal Research Center (CR-CHUM), Montreal, QC, Canada
| | - Chad M Vanderbilt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pooja Gogia
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Antoine Desilets
- Department of Hematology-Oncology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | | | - Daniel Kelly
- Informatics Systems, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew D Hellmann
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Laurence Zitvogel
- INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Charles M Rudin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Bertrand Routy
- University of Montreal Research Center (CR-CHUM), Montreal, QC, Canada
- Department of Hematology-Oncology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Lisa Derosa
- INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Adam J Schoenfeld
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
25
|
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
|
26
|
Mager LF, Krause T, McCoy KD. Interaction of microbiota, mucosal malignancies, and immunotherapy-Mechanistic insights. Mucosal Immunol 2024; 17:402-415. [PMID: 38521413 DOI: 10.1016/j.mucimm.2024.03.007] [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: 12/29/2023] [Revised: 03/09/2024] [Accepted: 03/17/2024] [Indexed: 03/25/2024]
Abstract
The microbiome has emerged as a crucial modulator of host-immune interactions and clearly impacts tumor development and therapy efficacy. The microbiome is a double-edged sword in cancer development and therapy as both pro-tumorigenic and anti-tumorigenic bacterial taxa have been identified. The staggering number of association-based studies in various tumor types has led to an enormous amount of data that makes it difficult to identify bacteria that promote tumor development or modulate therapy efficacy from bystander bacteria. Here we aim to comprehensively summarize the current knowledge of microbiome-host immunity interactions and cancer therapy in various mucosal tissues to find commonalities and thus identify potential functionally relevant bacterial taxa. Moreover, we also review recent studies identifying specific bacteria and mechanisms through which the microbiome modulates cancer development and therapy efficacy.
Collapse
Affiliation(s)
- Lukas F Mager
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Canada; Department of Internal Medicine I, Faculty of Medicine, University of Tübingen, Germany; M3 Research Center for Malignom, Metabolome and Microbiome, Faculty of Medicine University Tübingen, Germany
| | - Tim Krause
- Department of Internal Medicine I, Faculty of Medicine, University of Tübingen, Germany; M3 Research Center for Malignom, Metabolome and Microbiome, Faculty of Medicine University Tübingen, Germany
| | - Kathy D McCoy
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Canada.
| |
Collapse
|
27
|
Blake SJ, Wolf Y, Boursi B, Lynn DJ. Role of the microbiota in response to and recovery from cancer therapy. Nat Rev Immunol 2024; 24:308-325. [PMID: 37932511 DOI: 10.1038/s41577-023-00951-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2023] [Indexed: 11/08/2023]
Abstract
Our understanding of how the microbiota affects the balance between response to and failure of cancer treatment by modulating the tumour microenvironment and systemic immune system has advanced rapidly in recent years. Microbiota-targeting interventions in patients with cancer are an area of intensive investigation. Promisingly, phase I-II clinical trials have shown that interventions such as faecal microbiota transplantation can overcome resistance to immune checkpoint blockade in patients with melanoma, improve therapeutic outcomes in treatment-naive patients and reduce therapy-induced immunotoxicities. Here, we synthesize the evidence showing that the microbiota is an important determinant of both cancer treatment efficacy and treatment-induced acute and long-term toxicity, and we discuss the complex and inter-related mechanisms involved. We also assess the potential of microbiota-targeting interventions, including bacterial engineering and phage therapy, to optimize the response to and recovery from cancer therapy.
Collapse
Affiliation(s)
- Stephen J Blake
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Yochai Wolf
- Ella Lemelbaum Institute for Immuno-oncology and Skin Cancer, Sheba Medical Center, Tel Hashomer, Israel
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ben Boursi
- School of Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Oncology, Sheba Medical Center, Tel Hashomer, Israel
- Center of Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA, USA
| | - David J Lynn
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.
| |
Collapse
|
28
|
Wang X, Xu T, Wu S, Li L, Cai X, Chen F, Yan Z. Candida albicans-myeloid cells-T lymphocytes axis in the tumor microenvironment of oral tumor-bearing mice. Cancer Lett 2024; 588:216814. [PMID: 38499264 DOI: 10.1016/j.canlet.2024.216814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 03/20/2024]
Abstract
Candida albicans (C. albicans) is associated with the development of oral cancer. Here, we report the altered tumor microenvironment in oral tumor-bearing mice caused by C. albicans infection. Single-cell RNA sequencing showed that C. albicans infection influenced the tumor microenvironment significantly. Specifically, C. albicans infection reduced the CD8+ T cells but increased the IL-17A+ CD4+ T cells and IL-17A+ γδ T cells in oral tumor. The neutralization of IL-17A or TCR γ/δ alleviated the tumor progression caused by C. albicans infection. Additionally, C. albicans infection promoted the infiltration of myeloid-derived suppressor cells (MDSCs) into tumor, especially polymorphonuclear (PMN)-MDSCs, which infiltration was reduced after the neutralization of CCL2. Thus, our findings reveal the myeloid cells-T lymphocytes axis in oral tumor microenvironment with C. albicans infection, which helps to understand the mechanisms for C. albicans promoting oral cancer from the perspective of immune microenvironment.
Collapse
Affiliation(s)
- Xu Wang
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, China; National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Tiansong Xu
- Central Laboratory, Peking University School and Hospital of Stomatology, China; National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Shuangshuang Wu
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, China; National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Linman Li
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, China; National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Xinjia Cai
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, China; National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Feng Chen
- Central Laboratory, Peking University School and Hospital of Stomatology, China; National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
| | - Zhimin Yan
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, China; National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
| |
Collapse
|
29
|
Monyók Á, Mansour B, Vadnay I, Makra N, Dunai ZA, Nemes-Nikodém É, Stercz B, Szabó D, Ostorházi E. Change in Tissue Microbiome and Related Human Beta Defensin Levels Induced by Antibiotic Use in Bladder Carcinoma. Int J Mol Sci 2024; 25:4562. [PMID: 38674148 PMCID: PMC11050017 DOI: 10.3390/ijms25084562] [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: 03/15/2024] [Revised: 04/12/2024] [Accepted: 04/21/2024] [Indexed: 04/28/2024] Open
Abstract
It is now generally accepted that the success of antitumor therapy can be impaired by concurrent antibiotic therapy, the presence of certain bacteria, and elevated defensin levels around the tumor tissue. The aim of our current investigation was to identify the underlying changes in microbiome and defensin levels in the tumor tissue induced by different antibiotics, as well as the duration of this modification. The microbiome of the tumor tissues was significantly different from that of healthy volunteers. Comparing only the tumor samples, no significant difference was confirmed between the untreated group and the group treated with antibiotics more than 3 months earlier. However, antibiotic treatment within 3 months of analysis resulted in a significantly modified microbiome composition. Irrespective of whether Fosfomycin, Fluoroquinolone or Beta-lactam treatment was used, the abundance of Bacteroides decreased, and Staphylococcus abundance increased. Large amounts of the genus Acinetobacter were observed in the Fluoroquinolone-treated group. Regardless of the antibiotic treatment, hBD1 expression of the tumor cells consistently doubled. The increase in hBD2 and hBD3 expression was the highest in the Beta-lactam treated group. Apparently, antibiotic treatment within 3 months of sample analysis induced microbiome changes and defensin expression levels, depending on the identity of the applied antibiotic.
Collapse
Affiliation(s)
- Ádám Monyók
- Department of Urology, Markhot Ferenc University Teaching Hospital, 3300 Eger, Hungary; (Á.M.); (B.M.)
| | - Bassel Mansour
- Department of Urology, Markhot Ferenc University Teaching Hospital, 3300 Eger, Hungary; (Á.M.); (B.M.)
| | - István Vadnay
- Department of Pathology, Markhot Ferenc University Teaching Hospital, 3300 Eger, Hungary; (I.V.); (D.S.)
| | - Nóra Makra
- Department of Medical Microbiology, Semmelweis University, 1085 Budapest, Hungary; (N.M.); (Z.A.D.); (É.N.-N.); (B.S.)
| | - Zsuzsanna A. Dunai
- Department of Medical Microbiology, Semmelweis University, 1085 Budapest, Hungary; (N.M.); (Z.A.D.); (É.N.-N.); (B.S.)
| | - Éva Nemes-Nikodém
- Department of Medical Microbiology, Semmelweis University, 1085 Budapest, Hungary; (N.M.); (Z.A.D.); (É.N.-N.); (B.S.)
| | - Balázs Stercz
- Department of Medical Microbiology, Semmelweis University, 1085 Budapest, Hungary; (N.M.); (Z.A.D.); (É.N.-N.); (B.S.)
| | - Dóra Szabó
- Department of Pathology, Markhot Ferenc University Teaching Hospital, 3300 Eger, Hungary; (I.V.); (D.S.)
- Neurosurgery and Neurointervention Clinic, Semmelweis University, 1085 Budapest, Hungary
| | - Eszter Ostorházi
- Department of Pathology, Markhot Ferenc University Teaching Hospital, 3300 Eger, Hungary; (I.V.); (D.S.)
- Department of Dermatology, Venereology and Dermatooncology, Semmelweis University, 1085 Budapest, Hungary
| |
Collapse
|
30
|
Kong D, Wu Y, Liu Q, Huang C, Wang T, Huang Z, Gao Y, Li Y, Guo H. Functional analysis and validation of oncodrive gene AP3S1 in ovarian cancer through filtering of mutation data from whole-exome sequencing. Eur J Med Res 2024; 29:231. [PMID: 38609993 PMCID: PMC11015698 DOI: 10.1186/s40001-024-01814-7] [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: 01/19/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND High-grade serous ovarian carcinoma (HGSOC) is the most aggressive and prevalent subtype of ovarian cancer and accounts for a significant portion of ovarian cancer-related deaths worldwide. Despite advancements in cancer treatment, the overall survival rate for HGSOC patients remains low, thus highlighting the urgent need for a deeper understanding of the molecular mechanisms driving tumorigenesis and for identifying potential therapeutic targets. Whole-exome sequencing (WES) has emerged as a powerful tool for identifying somatic mutations and alterations across the entire exome, thus providing valuable insights into the genetic drivers and molecular pathways underlying cancer development and progression. METHODS Via the analysis of whole-exome sequencing results of tumor samples from 90 ovarian cancer patients, we compared the mutational landscape of ovarian cancer patients with that of TCGA patients to identify similarities and differences. The sequencing data were subjected to bioinformatics analysis to explore tumor driver genes and their functional roles. Furthermore, we conducted basic medical experiments to validate the results obtained from the bioinformatics analysis. RESULTS Whole-exome sequencing revealed the mutational profile of HGSOC, including BRCA1, BRCA2 and TP53 mutations. AP3S1 emerged as the most weighted tumor driver gene. Further analysis of AP3S1 mutations and expression demonstrated their associations with patient survival and the tumor immune response. AP3S1 knockdown experiments in ovarian cancer cells demonstrated its regulatory role in tumor cell migration and invasion through the TGF-β/SMAD pathway. CONCLUSION This comprehensive analysis of somatic mutations in HGSOC provides insight into potential therapeutic targets and molecular pathways for targeted interventions. AP3S1 was identified as being a key player in tumor immunity and prognosis, thus providing new perspectives for personalized treatment strategies. The findings of this study contribute to the understanding of HGSOC pathogenesis and provide a foundation for improved outcomes in patients with this aggressive disease.
Collapse
Affiliation(s)
- Deshui Kong
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 Huayuanbei Rd., Haidian District, Beijing, 100191, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, China
| | - Yu Wu
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 Huayuanbei Rd., Haidian District, Beijing, 100191, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, China
| | - Qiyu Liu
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 Huayuanbei Rd., Haidian District, Beijing, 100191, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, China
| | - Cuiyu Huang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 Huayuanbei Rd., Haidian District, Beijing, 100191, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, China
| | - Tongxia Wang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 Huayuanbei Rd., Haidian District, Beijing, 100191, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, China
| | - Zongyao Huang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 Huayuanbei Rd., Haidian District, Beijing, 100191, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, China
| | - Yan Gao
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 Huayuanbei Rd., Haidian District, Beijing, 100191, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, China
| | - Yuan Li
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 Huayuanbei Rd., Haidian District, Beijing, 100191, People's Republic of China.
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, China.
| | - Hongyan Guo
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 Huayuanbei Rd., Haidian District, Beijing, 100191, People's Republic of China.
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital), Beijing, China.
| |
Collapse
|
31
|
Zitvogel L, Kroemer G. Cancer and the Metaorganism. Cancer Discov 2024; 14:658-662. [PMID: 38571436 DOI: 10.1158/2159-8290.cd-23-1484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
SUMMARY Pathogenic shifts in the gut microbiota are part of the "ecological" alterations that accompany tumor progression and compromise immunosurveillance. The future management of health and disease including cancer will rely on the diagnosis of such shifts and their therapeutic correction by general or personalized strategies, hence restoring metaorganismal homeostasis.
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, Equipe 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
| | - 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
|
32
|
Luo WC, Mei SQ, Huang ZJ, Chen ZH, Zhang YC, Yang MY, Liu JQ, Xu JY, Yang XR, Zhong RW, Tang LB, Yin LX, Deng Y, Peng YL, Lu C, Chen BL, Ke DX, Tu HY, Yang JJ, Xu CR, Wu YL, Zhou Q. Correlation of distribution characteristics and dynamic changes of gut microbiota with the efficacy of immunotherapy in EGFR-mutated non-small cell lung cancer. J Transl Med 2024; 22:326. [PMID: 38566102 PMCID: PMC10985957 DOI: 10.1186/s12967-024-05135-5] [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: 02/04/2024] [Accepted: 03/27/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND The effects of gut microbiota and metabolites on the responses to immune checkpoint inhibitors (ICIs) in advanced epidermal growth factor receptor (EGFR) wild-type non-small cell lung cancer (NSCLC) have been studied. However, their effects on EGFR-mutated (EGFR +) NSCLC remain unknown. METHODS We prospectively recorded the clinicopathological characteristics of patients with advanced EGFR + NSCLC and assessed potential associations between the use of antibiotics or probiotics and immunotherapy efficacy. Fecal samples were collected at baseline, early on-treatment, response and progression status and were subjected to metagenomic next-generation sequencing and ultra-high-performance liquid chromatography-mass spectrometry analyses to assess the effects of gut microbiota and metabolites on immunotherapy efficacy. RESULTS The clinical data of 74 advanced EGFR + NSCLC patients were complete and 18 patients' fecal samples were dynamically collected. Patients that used antibiotics had shorter progression-free survival (PFS) (mPFS, 4.8 vs. 6.7 months; P = 0.037); probiotics had no impact on PFS. Two dynamic types of gut microbiota during immunotherapy were identified: one type showed the lowest relative abundance at the response time point, whereas the other type showed the highest abundance at the response time point. Metabolomics revealed significant differences in metabolites distribution between responders and non-responders. Deoxycholic acid, glycerol, and quinolinic acid were enriched in responders, whereas L-citrulline was enriched in non-responders. There was a significant correlation between gut microbiota and metabolites. CONCLUSIONS The use of antibiotics weakens immunotherapy efficacy in patients with advanced EGFR + NSCLC. The distribution characteristics and dynamic changes of gut microbiota and metabolites may indicate the efficacy of immunotherapy in advanced EGFR + NSCLC.
Collapse
Affiliation(s)
- Wei-Chi Luo
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Shi-Qi Mei
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Zi-Jian Huang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Zhi-Hong Chen
- Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yi-Chen Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Ming-Yi Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Jia-Qi Liu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Jing-Yan Xu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Xiao-Rong Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Ri-Wei Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Li-Bo Tang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Lin-Xi Yin
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yu Deng
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Ying-Long Peng
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Chang Lu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Bao-Long Chen
- Xiamen Treatgut Biotechnology Co., Ltd, Xiamen, China
| | - Dong-Xian Ke
- Xiamen Treatgut Medical Laboratory Co., Ltd, Xiamen, China
| | - Hai-Yan Tu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Jin-Ji Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Chong-Rui Xu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| |
Collapse
|
33
|
Li L, Chandra V, McAllister F. Tumor-resident microbes: the new kids on the microenvironment block. Trends Cancer 2024; 10:347-355. [PMID: 38388213 PMCID: PMC11006566 DOI: 10.1016/j.trecan.2023.12.002] [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] [Received: 07/17/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 02/24/2024]
Abstract
Tumor-resident microbes (TRM) are an integral component of the tumor microenvironment (TME). TRM can influence tumor growth, distant dissemination, and response to therapies by interfering with molecular pathways in tumor cells as well as with other components of the TME. Novel technologies are improving the identification and visualization of cell type-specific microbes in the TME. The mechanisms that mediate the role of TRM at the primary tumors and metastatic sites are being elucidated. This knowledge is providing novel perspectives for targeting microbes or using microbial interventions for cancer interception or therapy.
Collapse
Affiliation(s)
- Le Li
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vidhi Chandra
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Florencia McAllister
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
34
|
Geng S, Guo P, Li X, Shi Y, Wang J, Cao M, Zhang Y, Zhang K, Li A, Song H, Zhang Z, Shi J, Liu J, Yang Y. Biomimetic Nanovehicle-Enabled Targeted Depletion of Intratumoral Fusobacterium nucleatum Synergizes with PD-L1 Blockade against Breast Cancer. ACS NANO 2024; 18:8971-8987. [PMID: 38497600 DOI: 10.1021/acsnano.3c12687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Immune checkpoint blockade (ICB) therapy has been approved for breast cancer (BC), but clinical response rates are limited. Recent studies have shown that commensal microbes colonize a variety of tumors and are closely related to the host immune system response. Here, we demonstrated that Fusobacterium nucleatum (F.n), which is prevalent in BC, creates an immunosuppressive tumor microenvironment (ITME) characterized by a high-influx of myeloid cells that hinders ICB therapy. Administering the antibiotic metronidazole in BC can deplete F.n and remodel the ITME. To prevent an imbalance in the systemic microbiota caused by antibiotic administration, we designed a biomimetic nanovehicle for on-site antibiotic delivery inspired by F.n homing to BC. Additionally, ferritin-nanocaged doxorubicin was coloaded into this nanovehicle, as immunogenic chemotherapy has shown potential for synergy with ICB. It has been demonstrated that this biomimetic nanovehicle can be precisely homed to BC and efficiently eliminate intratumoral F.n without disrupting the diversity and abundance of systemic microbiota. This ultimately remodels the ITME, improving the therapeutic efficacy of the PD-L1 blocker with a tumor inhibition rate of over 90% and significantly extending the median survival of 4T1 tumor-bearing mice.
Collapse
Affiliation(s)
- Shizhen Geng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Pengke Guo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xinling Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yaru Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jing Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Mengnian Cao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yunya Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Airong Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Haiwei Song
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Junjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yiling Yang
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
35
|
López-Otín C, Kroemer G. The missing hallmark of health: psychosocial adaptation. Cell Stress 2024; 8:21-50. [PMID: 38476764 PMCID: PMC10928495 DOI: 10.15698/cst2024.03.294] [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: 01/11/2024] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 03/14/2024] Open
Abstract
The eight biological hallmarks of health that we initially postulated (Cell. 2021 Jan 7;184(1):33-63) include features of spatial compartmentalization (integrity of barriers, containment of local perturbations), maintenance of homeostasis over time (recycling & turnover, integration of circuitries, rhythmic oscillations) and an array of adequate responses to stress (homeostatic resilience, hormetic regulation, repair & regeneration). These hallmarks affect all eight somatic strata of the human body (molecules, organelles, cells, supracellular units, organs, organ systems, systemic circuitries and meta-organism). Here we postulate that mental and socioeconomic factors must be added to this 8×8 matrix as an additional hallmark of health ("psychosocial adaptation") and as an additional stratum ("psychosocial interactions"), hence building a 9×9 matrix. Potentially, perturbation of each of the somatic hallmarks and strata affects psychosocial factors and vice versa. Finally, we discuss the (patho)physiological bases of these interactions and their implications for mental health improvement.
Collapse
Affiliation(s)
- Carlos López-Otín
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Facultad de Ciencias de la Vida y la Naturaleza, Universidad Nebrija, Madrid, Spain
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| |
Collapse
|
36
|
Kroemer G, Chan TA, Eggermont AMM, Galluzzi L. Immunosurveillance in clinical cancer management. CA Cancer J Clin 2024; 74:187-202. [PMID: 37880100 PMCID: PMC10939974 DOI: 10.3322/caac.21818] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/27/2023] [Accepted: 10/03/2023] [Indexed: 10/27/2023] Open
Abstract
The progression of cancer involves a critical step in which malignant cells escape from control by the immune system. Antineoplastic agents are particularly efficient when they succeed in restoring such control (immunosurveillance) or at least establish an equilibrium state that slows down disease progression. This is true not only for immunotherapies, such as immune checkpoint inhibitors (ICIs), but also for conventional chemotherapy, targeted anticancer agents, and radiation therapy. Thus, therapeutics that stress and kill cancer cells while provoking a tumor-targeting immune response, referred to as immunogenic cell death, are particularly useful in combination with ICIs. Modern oncology regimens are increasingly using such combinations, which are referred to as chemoimmunotherapy, as well as combinations of multiple ICIs. However, the latter are generally associated with severe side effects compared with single-agent ICIs. Of note, the success of these combinatorial strategies against locally advanced or metastatic cancers is now spurring successful attempts to move them past the postoperative (adjuvant) setting to the preoperative (neoadjuvant) setting, even for patients with operable cancers. Here, the authors critically discuss the importance of immunosurveillance in modern clinical cancer management.
Collapse
Affiliation(s)
- Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France; Institut du Cancer Paris Carpem, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Timothy A. Chan
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA; Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; National Center for Regenerative Medicine, Cleveland, OH, USA; Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Alexander M. M. Eggermont
- University Medical Center Utrecht & Princess Maxima Center, Utrecht, the Netherlands; Comprehensive Cancer Center München, Technical University München & Ludwig Maximilian University, München, Germany
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| |
Collapse
|
37
|
Alves Costa Silva C, Piccinno G, Suissa D, Bourgin M, Schreibelt G, Durand S, Birebent R, Fidelle M, Sow C, Aprahamian F, Manghi P, Punčochář M, Asnicar F, Pinto F, Armanini F, Terrisse S, Routy B, Drubay D, Eggermont AMM, Kroemer G, Segata N, Zitvogel L, Derosa L, Bol KF, de Vries IJM. Influence of microbiota-associated metabolic reprogramming on clinical outcome in patients with melanoma from the randomized adjuvant dendritic cell-based MIND-DC trial. Nat Commun 2024; 15:1633. [PMID: 38395948 PMCID: PMC10891084 DOI: 10.1038/s41467-024-45357-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Tumor immunosurveillance plays a major role in melanoma, prompting the development of immunotherapy strategies. The gut microbiota composition, influencing peripheral and tumoral immune tonus, earned its credentials among predictors of survival in melanoma. The MIND-DC phase III trial (NCT02993315) randomized (2:1 ratio) 148 patients with stage IIIB/C melanoma to adjuvant treatment with autologous natural dendritic cell (nDC) or placebo (PL). Overall, 144 patients collected serum and stool samples before and after 2 bimonthly injections to perform metabolomics (MB) and metagenomics (MG) as prespecified exploratory analysis. Clinical outcomes are reported separately. Here we show that different microbes were associated with prognosis, with the health-related Faecalibacterium prausnitzii standing out as the main beneficial taxon for no recurrence at 2 years (p = 0.008 at baseline, nDC arm). Therapy coincided with major MB perturbations (acylcarnitines, carboxylic and fatty acids). Despite randomization, nDC arm exhibited MG and MB bias at baseline: relative under-representation of F. prausnitzii, and perturbations of primary biliary acids (BA). F. prausnitzii anticorrelated with BA, medium- and long-chain acylcarnitines. Combined, these MG and MB biomarkers markedly determined prognosis. Altogether, the host-microbial interaction may play a role in localized melanoma. We value systematic MG and MB profiling in randomized trials to avoid baseline differences attributed to host-microbe interactions.
Collapse
Grants
- The MIND-DC trial was funded by ZonMw, Ministry of Health, Welfare and Sport (VWS), Stichting ATK, Miltenyi Biotec (in-kind). This work was supported by SEERAVE Foundation, European Union Horizon 2020:Project Number: 825410 and Project Acronym: ONCOBIOME, Institut National du Cancer (INCa), ANR Ileobiome - 19-CE15-0029-01, ANR RHU5 “ANR-21-RHUS-0017” IMMUNOLIFE&#x201D;, MAdCAM INCA_ 16698, Ligue contre le cancer, LABEX OncoImmunology, la direction generale de l&#x2019;offre de soins (DGOS), Universite Paris-Sud, SIRIC SOCRATE (INCa/DGOS/INSERM 6043), and PACRI network. G.K. is supported by the Ligue contre le Cancer (équipe labellis&#x00E9;e); Agence National de la Recherche (ANR) – Projets blancs; AMMICa US23/CNRS UMS3655; Association pour la recherche sur le cancer (ARC); Canc&#x00E9;rop&#x00F4;le Ile-de-France; Fondation pour la Recherche M&#x00E9;dicale (FRM); a donation by Elior; Equipex Onco-Pheno-Screen; European Joint Programme on Rare Diseases (EJPRD); European Research Council Advanced Investigator Award (ERC-2021-ADG, ICD-Cancer, Grant No. 101052444), European Union Horizon 2020 Projects Oncobiome, Prevalung (grant No. 101095604) and Crimson; Fondation Carrefour; Institut National du Cancer (INCa); Institut Universitaire de France; LabEx Immuno-Oncology (ANR-18-IDEX-0001); a Cancer Research ASPIRE Award from the Mark Foundation; the RHU Immunolife; Seerave Foundation; SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination (SOCRATE); and SIRIC Cancer Research and Personalized Medicine (CARPEM). This study contributes to the IdEx Universit&#x00E9; de Paris ANR-18-IDEX-0001. This work is supported by the Prism project funded by the Agence Nationale de la Recherche under grant number ANR-18-IBHU-0002. CACS was funded by MSD Avenir. MF is funded by SEERAVE Foundation and MERCK Foundation. LD and BR were supported by Philantropia at Gustave Roussy Foundation.
Collapse
Affiliation(s)
- Carolina Alves Costa Silva
- Gustave Roussy Cancer Campus (GRCC), ClinicObiome, Villejuif Cedex, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Gianmarco Piccinno
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Déborah Suissa
- Gustave Roussy Cancer Campus (GRCC), ClinicObiome, Villejuif Cedex, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Mélanie Bourgin
- Metabolomics and Cell Biology Platforms, 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
| | - Gerty Schreibelt
- Medical BioSciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Sylvère Durand
- Metabolomics and Cell Biology Platforms, 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
| | - Roxanne Birebent
- Gustave Roussy Cancer Campus (GRCC), ClinicObiome, Villejuif Cedex, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Marine Fidelle
- Gustave Roussy Cancer Campus (GRCC), ClinicObiome, Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Cissé Sow
- Gustave Roussy Cancer Campus (GRCC), ClinicObiome, Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Fanny Aprahamian
- Metabolomics and Cell Biology Platforms, 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
| | - Paolo Manghi
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Michal Punčochář
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Francesco Asnicar
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Federica Pinto
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Federica Armanini
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Safae Terrisse
- Oncology Department, Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis, Paris, France
| | - Bertrand Routy
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Hematology-Oncology Division, Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | - Damien Drubay
- Gustave Roussy Cancer Campus (GRCC), ClinicObiome, Villejuif Cedex, France
- Office of Biostatistics and Epidemiology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
- Inserm, Université Paris-Saclay, CESP U1018, Oncostat, labeled Ligue Contre le Cancer, Villejuif, France
| | - Alexander M M Eggermont
- Princess Máxima Center and University Medical Center Utrecht, 3584 CS Utrecht, The Netherlands
- Comprehensive Cancer Center Munich, Technical University Munich & Ludwig Maximiliaan University, Munich, Germany
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, 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
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Nicola Segata
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, Trento, Italy
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus (GRCC), ClinicObiome, Villejuif Cedex, France.
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France.
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France.
- Center of Clinical Investigations BIOTHERIS, INSERM CIC1428, Villejuif, France.
| | - Lisa Derosa
- Gustave Roussy Cancer Campus (GRCC), ClinicObiome, Villejuif Cedex, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Kalijn F Bol
- Medical BioSciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
- Department of Medical Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Medical BioSciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
38
|
Sundström P, Hogg S, Quiding Järbrink M, Bexe Lindskog E. Immune cell infiltrates in peritoneal metastases from colorectal cancer. Front Immunol 2024; 15:1347900. [PMID: 38384469 PMCID: PMC10879551 DOI: 10.3389/fimmu.2024.1347900] [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: 12/01/2023] [Accepted: 01/24/2024] [Indexed: 02/23/2024] Open
Abstract
Background The presence of peritoneal metastases (PMs) in patients with colorectal cancer (CRC) confers a poor prognosis and only a minority of patients will benefit from the available treatment options. In primary CRC tumors, it is well established that a high infiltration of CD8+ effector T cells correlates to a favorable patient outcome. In contrast, the immune response induced in PMs from CRC and how it relates to patient survival is still unknown. In this study, we characterized the immune infiltrates and the distribution of immune checkpoint receptors on T cells from PMs from CRC, in order to evaluate the potential benefit of checkpoint blockade immunotherapy for this patient group. Methods Surgically resected PM tissue from CRC patients (n=22) and synchronous primary tumors (n=8) were processed fresh to single cell suspensions using enzymatic digestion. Surface markers and cytokine production were analyzed using flow cytometry. Results T cells dominated the leukocyte infiltrate in the PM specimens analyzed, followed by monocytes and B cells. Comparing two different PMs from the same patient usually showed a similar distribution of immune cells in both samples. The T cell infiltrate was characterized by an activated phenotype and markers of exhaustion were enriched compared with matched circulating T cells, in particular the checkpoint receptors PD-1 and TIGIT. In functional assays most cytotoxic and helper T cells produced INF-γ and TNF following polyclonal stimulation, while few produced IL-17, indicating a dominance of Th1-type responses in the microenvironment of PMs. Conclusion Immune cells were present in all PMs from CRC examined. Although infiltrating T cells express markers of exhaustion, they produce Th1-type cytokines when stimulated. These results indicate the possibility to augment tumor-specific immune responses within PMs using checkpoint blockade inhibitors.
Collapse
Affiliation(s)
- Patrik Sundström
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Stephen Hogg
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Marianne Quiding Järbrink
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Elinor Bexe Lindskog
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
39
|
Xin Y, Liu CG, Zang D, Chen J. Gut microbiota and dietary intervention: affecting immunotherapy efficacy in non-small cell lung cancer. Front Immunol 2024; 15:1343450. [PMID: 38361936 PMCID: PMC10867196 DOI: 10.3389/fimmu.2024.1343450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/18/2024] [Indexed: 02/17/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for 80-85% of all lung cancers. In recent years, treatment with immune checkpoint inhibitors (ICIs) has gradually improved the survival rate of patients with NSCLC, especially those in the advanced stages. ICIs can block the tolerance pathways that are overexpressed by tumor cells and maintain the protective activity of immune system components against cancer cells. Emerging clinical evidence suggests that gut microbiota may modulate responses to ICIs treatment, possibly holding a key role in tumor immune surveillance and the efficacy of ICIs. Studies have also shown that diet can influence the abundance of gut microbiota in humans, therefore, dietary interventions and the adjustment of the gut microbiota is a novel and promising treatment strategy for adjunctive cancer therapy. This review comprehensively summarizes the effects of gut microbiota, antibiotics (ATBs), and dietary intervention on the efficacy of immunotherapy in NSCLC, with the aim of informing the development of novel strategies in NSCLC immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Jun Chen
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
40
|
Routy B, Jackson T, Mählmann L, Baumgartner CK, Blaser M, Byrd A, Corvaia N, Couts K, Davar D, Derosa L, Hang HC, Hospers G, Isaksen M, Kroemer G, Malard F, McCoy KD, Meisel M, Pal S, Ronai Z, Segal E, Sepich-Poore GD, Shaikh F, Sweis RF, Trinchieri G, van den Brink M, Weersma RK, Whiteson K, Zhao L, McQuade J, Zarour H, Zitvogel L. Melanoma and microbiota: Current understanding and future directions. Cancer Cell 2024; 42:16-34. [PMID: 38157864 PMCID: PMC11096984 DOI: 10.1016/j.ccell.2023.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 01/03/2024]
Abstract
Over the last decade, the composition of the gut microbiota has been found to correlate with the outcomes of cancer patients treated with immunotherapy. Accumulating evidence points to the various mechanisms by which intestinal bacteria act on distal tumors and how to harness this complex ecosystem to circumvent primary resistance to immune checkpoint inhibitors. Here, we review the state of the microbiota field in the context of melanoma, the recent breakthroughs in defining microbial modes of action, and how to modulate the microbiota to enhance response to cancer immunotherapy. The host-microbe interaction may be deciphered by the use of "omics" technologies, and will guide patient stratification and the development of microbiota-centered interventions. Efforts needed to advance the field and current gaps of knowledge are also discussed.
Collapse
Affiliation(s)
- Bertrand Routy
- University of Montreal Research Center (CRCHUM), Montreal, QC H2X 0A9, Canada; Hematology-Oncology Division, Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC H2X 3E4, Canada
| | - Tanisha Jackson
- Melanoma Research Alliance, 730 15th Street NW, Washington, DC 20005, USA
| | - Laura Mählmann
- Seerave Foundation, The Seerave Foundation, 35-37 New Street, St Helier, JE2 3RA Jersey, UK
| | | | - Martin Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ 08854, USA
| | - Allyson Byrd
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA 94080, USA
| | | | - Kasey Couts
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Diwakar Davar
- Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lisa Derosa
- Gustave Roussy Cancer Center, ClinicoBiome, 94805 Villejuif, France; Université Paris Saclay, Faculty of Medicine, 94270 Kremlin Bicêtre, France; Inserm U1015, Equipe Labellisée par la Ligue Contre le Cancer, 94800 Villejuif, France
| | - Howard C Hang
- Departments of Immunology & Microbiology and Chemistry, Scripps Research, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Geke Hospers
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, The Netherlands
| | | | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France; Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94905 Villejuif, France; Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France
| | - Florent Malard
- Sorbonne Université, Centre de Recherche Saint-Antoine INSERM UMRs938, Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, AP-HP, Paris, France
| | - Kathy D McCoy
- Department of Physiology & Pharmacology, Snyder Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Marlies Meisel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA USA
| | - Sumanta Pal
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Ze'ev Ronai
- Sanford Burnham Prebys Discovery Medical Research Institute, La Jolla, CA 92037, USA
| | - Eran Segal
- Weizmann Institute of Science, Computer Science and Applied Mathematics Department, 234th Herzel st., Rehovot 7610001, Israel
| | - Gregory D Sepich-Poore
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Micronoma Inc., San Diego, CA 92121, USA
| | - Fyza Shaikh
- Johns Hopkins School of Medicine, Department of Oncology, Baltimore, MD 21287, USA
| | - Randy F Sweis
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Giorgio Trinchieri
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marcel van den Brink
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Immunology, Sloan Kettering Institute, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Katrine Whiteson
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Liping Zhao
- Department of Biochemistry and Microbiology, New Jersey Institute of Food, Nutrition and Health, Rutgers University, New Brunswick, NY 08901, USA
| | - Jennifer McQuade
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Hassane Zarour
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA.
| | - Laurence Zitvogel
- Gustave Roussy Cancer Center, ClinicoBiome, 94805 Villejuif, France; Université Paris Saclay, Faculty of Medicine, 94270 Kremlin Bicêtre, France; Inserm U1015, Equipe Labellisée par la Ligue Contre le Cancer, 94800 Villejuif, France; Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Gustave Roussy, 94805 Villejuif, France.
| |
Collapse
|
41
|
Galván-Peña S, Zhu Y, Hanna BS, Mathis D, Benoist C. A dynamic atlas of immunocyte migration from the gut. Sci Immunol 2024; 9:eadi0672. [PMID: 38181094 PMCID: PMC10964343 DOI: 10.1126/sciimmunol.adi0672] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 12/06/2023] [Indexed: 01/07/2024]
Abstract
Dysbiosis in the gut microbiota affects several systemic diseases, possibly by driving the migration of perturbed intestinal immunocytes to extraintestinal tissues. Combining Kaede photoconvertible mice and single-cell genomics, we generated a detailed map of migratory trajectories from the colon, at baseline, and in several models of intestinal and extraintestinal inflammation. All lineages emigrated from the colon in an S1P-dependent manner. B lymphocytes represented the largest contingent, with the unexpected circulation of nonexperienced follicular B cells, which carried a gut-imprinted transcriptomic signature. T cell emigration included distinct groups of RORγ+ and IEL-like CD160+ subsets. Gut inflammation curtailed emigration, except for dendritic cells disseminating to lymph nodes. Colon-emigrating cells distributed differentially to distinct sites of extraintestinal models of inflammation (psoriasis-like skin, arthritic synovium, and tumors). Thus, specific cellular trails originating in the gut and influenced by microbiota may shape peripheral immunity in varied ways.
Collapse
Affiliation(s)
| | - Yangyang Zhu
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Bola S. Hanna
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
42
|
Bernier-Latmani J, González-Loyola A, Petrova TV. Mechanisms and functions of intestinal vascular specialization. J Exp Med 2024; 221:e20222008. [PMID: 38051275 PMCID: PMC10697212 DOI: 10.1084/jem.20222008] [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: 09/11/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023] Open
Abstract
The intestinal vasculature has been studied for the last 100 years, and its essential role in absorbing and distributing ingested nutrients is well known. Recently, fascinating new insights into the organization, molecular mechanisms, and functions of intestinal vessels have emerged. These include maintenance of intestinal epithelial cell function, coping with microbiota-induced inflammatory pressure, recruiting gut-specific immune cells, and crosstalk with other organs. Intestinal function is also regulated at the systemic and cellular levels, such that the postprandial hyperemic response can direct up to 30% of systemic blood to gut vessels, while micron-sized endothelial cell fenestrations are necessary for nutrient uptake. In this review, we will highlight past discoveries made about intestinal vasculature in the context of new findings of molecular mechanisms underpinning gut function. Such comprehensive understanding of the system will pave the way to breakthroughs in nutrient uptake optimization, drug delivery efficiency, and treatment of human diseases.
Collapse
Affiliation(s)
- Jeremiah Bernier-Latmani
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | | | - Tatiana V. Petrova
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, École polytechnique fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
43
|
Paz Del Socorro T, Oka K, Boulard O, Takahashi M, Poulin LF, Hayashi A, Chamaillard M. The biotherapeutic Clostridium butyricum MIYAIRI 588 strain potentiates enterotropism of Rorγt +Treg and PD-1 blockade efficacy. Gut Microbes 2024; 16:2315631. [PMID: 38385162 PMCID: PMC10885180 DOI: 10.1080/19490976.2024.2315631] [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: 09/20/2023] [Accepted: 02/02/2024] [Indexed: 02/23/2024] Open
Abstract
Immune checkpoint inhibitors (ICI) have been positioned as a standard of care for patients with advanced non-small-cell lung carcinomas (NSCLC). A pilot clinical trial has reflected optimistic association between supplementation with Clostridium butyricum MIYAIRI 588 (CBM588) and ICI efficacy in NSCLC. However, it remains to be established whether this biotherapeutic strain may be sufficient to heighten the immunogenicity of the tumor draining lymph nodes to overcome resistance to ICI. Herein, we report that supplementation with CBM588 led to an improved responsiveness to antibody targeting programmed cell death protein 1 (aPD-1). This was statistically associated with a significant decrease in α-diversity of gut microbiota from CBM588-treated mice upon PD-1 blockade. At the level of the tumor-draining lymph node, such combination of treatment significantly lowered the frequency of microbiota-modulated subset of regulatory T cells that express Retinoic Orphan Receptor gamma t (Rorγ t+ Treg). Specifically, this strongly immunosuppressive was negatively correlated with the abundance of bacteria that belong to the family of Ruminococcaceae. Accordingly, the colonic expression of both indoleamine 2,3-Dioxygenase 1 (IDO-1) and interleukin-10 (IL-10) were heightened in mice with greater PD-1 blockade efficacy. The CBM588-induced ability to secrete Interleukin-10 of lamina propria mononuclear cells was heightened in tumor bearers when compared with cancer-free mice. Conversely, blockade of interleukin-10 signaling preferentially enhanced the capacity of CD8+ T cells to secrete Interferon gamma when being cocultured with CBM588-primed lamina propria mononuclear cells of tumor-bearing mice. Our results demonstrate that CBM588-centered intervention can adequately improve intestinal homeostasis and efficiently overcome resistance to PD-1 blockade in mice.
Collapse
Affiliation(s)
| | - Kentaro Oka
- R&D Division, Miyarisan Pharmaceutical Co., Ltd, Saitama, Japan
| | | | | | | | - Atsushi Hayashi
- R&D Division, Miyarisan Pharmaceutical Co., Ltd, Saitama, Japan
| | | |
Collapse
|
44
|
Lee PJ, Hung CM, Yang AJ, Hou CY, Chou HW, Chang YC, Chu WC, Huang WY, Kuo WC, Yang CC, Lin KI, Hung KH, Chang LC, Lee KY, Kuo HP, Lu KM, Lai HC, Kuo ML, Chen WJ. MS-20 enhances the gut microbiota-associated antitumor effects of anti-PD1 antibody. Gut Microbes 2024; 16:2380061. [PMID: 39078050 PMCID: PMC11290773 DOI: 10.1080/19490976.2024.2380061] [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: 03/10/2024] [Revised: 06/28/2024] [Accepted: 07/10/2024] [Indexed: 07/31/2024] Open
Abstract
Cancer immunotherapy has been regarded as a promising strategy for cancer therapy by blocking immune checkpoints and evoking immunity to fight cancer, but its efficacy seems to be heterogeneous among patients. Manipulating the gut microbiota is a potential strategy for enhancing the efficacy of immunotherapy. Here, we report that MS-20, also known as "Symbiota®", a postbiotic that comprises abundant microbial metabolites generated from a soybean-based medium fermented with multiple strains of probiotics and yeast, inhibited colon and lung cancer growth in combination with an anti-programmed cell death 1 (PD1) antibody in xenograft mouse models. Mechanistically, MS-20 remodeled the immunological tumor microenvironment by increasing effector CD8+ T cells and downregulating PD1 expression, which were mediated by the gut microbiota. Fecal microbiota transplantation (FMT) from mice receiving MS-20 treatment to recipient mice increased CD8+ T-cell infiltration into the tumor microenvironment and significantly improved antitumor activity when combined with anti-PD1 therapy. Notably, the abundance of Ruminococcus bromii, which increased following MS-20 treatment, was positively associated with a reduced tumor burden and CD8+ T-cell infiltration in vivo. Furthermore, an ex vivo study revealed that MS-20 could alter the composition of the microbiota in cancer patients, resulting in distinct metabolic pathways associated with favorable responses to immunotherapy. Overall, MS-20 could act as a promising adjuvant agent for enhancing the efficacy of immune checkpoint-mediated antitumor therapy.
Collapse
Affiliation(s)
- Pei-Jung Lee
- Pharmaceutical Research & Development, Microbio Co, Ltd, Taipei, Taiwan
| | - Chien-Min Hung
- Pharmaceutical Research & Development, Microbio Co, Ltd, Taipei, Taiwan
| | - Ai-Jen Yang
- Pharmaceutical Research & Development, Microbio Co, Ltd, Taipei, Taiwan
| | - Cheng-Yu Hou
- Pharmaceutical Research & Development, Microbio Co, Ltd, Taipei, Taiwan
| | - Hung-Wen Chou
- Animal Center for Drug Screening, Oneness Biotech Co, Ltd, Taipei, Taiwan
| | - Yi-Chung Chang
- Nucleic Acid Drug Division, Microbio (Shanghai) Biotech Company, Shanghai, China
| | - Wen-Cheng Chu
- Pharmaceutical Research & Development, Microbio Co, Ltd, Taipei, Taiwan
| | - Wen-Yen Huang
- Pharmaceutical Research & Development, Microbio Co, Ltd, Taipei, Taiwan
| | - Wen-Chih Kuo
- Pharmaceutical Research & Development, Microbio Co, Ltd, Taipei, Taiwan
| | - Chia-Chun Yang
- Nucleic Acid Drug Division, Microbio (Shanghai) Biotech Company, Shanghai, China
| | - Kuo-I Lin
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Kuo-Hsuan Hung
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Li-Chun Chang
- Division of Gastroenterology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Han-Pin Kuo
- Pulmonary Medicine Research Center, Taipei Medical University, Taipei, Taiwan
- Department of Thoracic Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Kung-Ming Lu
- General manager’s office, Microbio Co., Ltd., Taipei, Taiwan
| | - Hsin-Chih Lai
- General manager’s office, Revivebio Co, Ltd, Taipei, Taiwan
| | - Ming-Liang Kuo
- General manager’s office, Microbio Co., Ltd., Taipei, Taiwan
| | - Wan-Jiun Chen
- Pharmaceutical Research & Development, Microbio Co, Ltd, Taipei, Taiwan
| |
Collapse
|
45
|
Abstract
The remarkable diversity of lymphocytes, essential components of the immune system, serves as an ingenious mechanism for maximizing the efficient utilization of limited host defense resources. While cell adhesion molecules, notably in gut-tropic T cells, play a central role in this mechanism, the counterbalancing molecular details have remained elusive. Conversely, we've uncovered the molecular pathways enabling extracellular vesicles secreted by lymphocytes to reach the gut's mucosal tissues, facilitating immunological regulation. This discovery sheds light on immune fine-tuning, offering insights into immune regulation mechanisms.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Ryu Yashiro
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Higashimurayama, Tokyo, Japan
| |
Collapse
|
46
|
Jiang Y, Jia D, Sun Y, Ding N, Wang L. Microbiota: A key factor affecting and regulating the efficacy of immunotherapy. Clin Transl Med 2023; 13:e1508. [PMID: 38082435 PMCID: PMC10713876 DOI: 10.1002/ctm2.1508] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Immunotherapy has made significant progress in cancer treatment; however, the responsiveness to immunotherapy varies widely among patients. Growing evidence has demonstrated the role of the gut microbiota in the efficacy of immunotherapy. MAIN BODY Herein, we summarise the changes in the microbiota in different cancers under various immunotherapies. The microbial-host signal transmission on immunotherapeutic responses and mechanisms associated with microbial translocation to tumours in the context of immunotherapy are also discussed. In addition, we have highlighted the clinical application value of methods for regulating the microbiota. Finally, we elaborate on the relationship between the microbiota, host and immunotherapy, and provide potential directions for future research. CONCLUSION Different microbiota cause changes in the tumour microenvironment through microbial signals thereby affecting immunotherapy efficacy. Translocation of gut microbiota and the role of extraintestinal microbiota in immunotherapy deserve attention. Microbiota regulation is a novel strategy for combination therapy with immunotherapy. Although there are several aspects that deserve further refinement and exploration with regard to administration and clinical translation. Nevertheless, it is foreseeable that the microbiota will become an integral part of cancer treatment.
Collapse
Affiliation(s)
- Yao Jiang
- Department of GastroenterologySecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institution of GastroenterologyZhejiang UniversityHangzhouChina
| | - Dingjiacheng Jia
- Department of GastroenterologySecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institution of GastroenterologyZhejiang UniversityHangzhouChina
| | - Yong Sun
- Department of GastroenterologySecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institution of GastroenterologyZhejiang UniversityHangzhouChina
| | - Ning Ding
- 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
|
47
|
Abbondio M, Tanca A, De Diego L, Sau R, Bibbò S, Pes GM, Dore MP, Uzzau S. Metaproteomic assessment of gut microbial and host functional perturbations in Helicobacter pylori-infected patients subjected to an antimicrobial protocol. Gut Microbes 2023; 15:2291170. [PMID: 38063474 PMCID: PMC10730194 DOI: 10.1080/19490976.2023.2291170] [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: 08/31/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
The impact of therapeutic interventions on the human gut microbiota (GM) is a clinical issue of paramount interest given the strong interconnection between microbial dynamics and human health. Orally administered antibiotics are known to reduce GM biomass and modify GM taxonomic profile. However, the impact of antimicrobial therapies on GM functions and biochemical pathways has scarcely been studied. Here, we characterized the fecal metaproteome of 10 Helicobacter pylori-infected patients before (T0) and after 10 days (T1) of a successful quadruple therapy (bismuth, tetracycline, metronidazole, and rabeprazole) and 30 days after therapy cessation (T2), to investigate how GM and host functions change during the eradication and healing processes. At T1, the abundance ratio between microbial and host proteins was reversed compared with that at T0 and T2. Several pathobionts (including Klebsiella, Proteus, Enterococcus, Muribaculum, and Enterocloster) were increased at T1. Therapy reshaped the relative contributions of the functions required to produce acetate, propionate, and butyrate. Proteins related to the uptake and processing of complex glycans were increased. Microbial cross-feeding with sialic acid, fucose, and rhamnose was enhanced, whereas hydrogen sulfide production was reduced. Finally, microbial proteins involved in antibiotic resistance and inflammation were more abundant after therapy. Moreover, a reduction in host proteins with known roles in inflammation and H. pylori-mediated carcinogenesis was observed. In conclusion, our results support the use of metaproteomics to monitor drug-induced remodeling of GM and host functions, opening the way for investigating new antimicrobial therapies aimed at preserving gut environmental homeostasis.
Collapse
Affiliation(s)
- Marcello Abbondio
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Alessandro Tanca
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Laura De Diego
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Rosangela Sau
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Stefano Bibbò
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Giovanni Mario Pes
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Maria Pina Dore
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Sergio Uzzau
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
48
|
Laurans L, Mouttoulingam N, Chajadine M, Lavelle A, Diedisheim M, Bacquer E, Creusot L, Suffee N, Esposito B, Melhem NJ, Le Goff W, Haddad Y, Paul JL, Rainteau D, Tedgui A, Ait-Oufella H, Zitvogel L, Sokol H, Taleb S. An obesogenic diet increases atherosclerosis through promoting microbiota dysbiosis-induced gut lymphocyte trafficking into the periphery. Cell Rep 2023; 42:113350. [PMID: 37897726 DOI: 10.1016/j.celrep.2023.113350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 09/13/2023] [Accepted: 10/11/2023] [Indexed: 10/30/2023] Open
Abstract
Although high-fat diet (HFD)-induced gut microbiota dysbiosis is known to affect atherosclerosis, the underlying mechanisms remain to be fully explored. Here, we show that the progression of atherosclerosis depends on a gut microbiota shaped by an HFD but not a high-cholesterol (HC) diet and, more particularly, on low fiber (LF) intake. Mechanistically, gut lymphoid cells impacted by HFD- or LF-induced microbiota dysbiosis highly proliferate in mesenteric lymph nodes (MLNs) and migrate from MLNs to the periphery, which fuels T cell accumulation within atherosclerotic plaques. This is associated with the induction of mucosal addressin cell adhesion molecule 1 (MAdCAM-1) within plaques and the presence of enterotropic lymphocytes expressing β7 integrin. MLN resection or lymphocyte deficiency abrogates the pro-atherogenic effects of a microbiota shaped by LF. Our study shows a pathological link between a diet-shaped microbiota, gut immune cells, and atherosclerosis, suggesting that a diet-modulated microbiome might be a suitable therapeutic target to prevent atherosclerosis.
Collapse
Affiliation(s)
- Ludivine Laurans
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Nirmala Mouttoulingam
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Mouna Chajadine
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Aonghus Lavelle
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, 75012 Paris, France; Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Marc Diedisheim
- Clinique Saint Gatien Alliance (NCT+), 37540 Saint-Cyr-sur-Loire, France; Institut Necker-Enfants Malades (INEM), Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, 75015 Paris, France
| | - Emilie Bacquer
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Laura Creusot
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, 75012 Paris, France; Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Nadine Suffee
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France; INSERM UMRS1166, ICAN-Institute of Cardiometabolism and Nutrition, Sorbonne University, 75013 Paris, France
| | - Bruno Esposito
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Nada Joe Melhem
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Wilfried Le Goff
- INSERM UMRS1166, ICAN-Institute of Cardiometabolism and Nutrition, Sorbonne University, 75013 Paris, France
| | - Yacine Haddad
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France; Gustave Roussy, Villejuif, France; Institut National de la Santé et de la Recherche Médicale, Gustave Roussy, UMR1015, Villejuif, France
| | - Jean-Louis Paul
- Université Paris-Sud, Equipe d'Accueil 4529, UFR de Pharmacie, Chatenay-Malabry, France and Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Dominique Rainteau
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France; Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Clinical Metabolomics Department, 75012 Paris, France
| | - Alain Tedgui
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Hafid Ait-Oufella
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Laurence Zitvogel
- Gustave Roussy, Villejuif, France; Institut National de la Santé et de la Recherche Médicale, Gustave Roussy, UMR1015, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France; Center of Clinical Investigations BIOTHERIS, INSERM CIC1428, Gustave Roussy, Villejuif, France
| | - Harry Sokol
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, 75012 Paris, France; Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France; INRAe, Micalis & AgroParisTech, Jouy en Josas, France
| | - Soraya Taleb
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France.
| |
Collapse
|
49
|
Zhang H, Xu Z. Gut-lung axis: role of the gut microbiota in non-small cell lung cancer immunotherapy. Front Oncol 2023; 13:1257515. [PMID: 38074650 PMCID: PMC10701269 DOI: 10.3389/fonc.2023.1257515] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/03/2023] [Indexed: 11/02/2024] Open
Abstract
Immunotherapy for non-small cell lung cancer (NSCLC) has advanced considerably over the past two decades. In particular, immune checkpoint inhibitors are widely used for treating NSCLC. However, the overall cure and survival rates of patients with NSCLC remain low. Therefore, continuous investigation into complementary treatments is necessary to expand the clinical advantages of immunotherapy to a larger cohort of patients with NSCLC. Recently, the distinctive role of the gut microbiota (GM) in the initiation, progression, and dissemination of cancer has attracted increasing attention. Emerging evidence indicates a close relationship between the gut and lungs, known as the gut-lung axis (GLA). In this review, we aim to provide a comprehensive summary of the current knowledge regarding the connection between the GM and the outcomes of immunotherapy in NSCLC, with particular focus on the recent understanding of GLA. Overall, promising GM-based therapeutic strategies have been observed to improve the effectiveness or reduce the toxicity of immunotherapy in patients with NSCLC, thus advancing the utilization of microbiota precision medicine.
Collapse
Affiliation(s)
- Huaiyuan Zhang
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ziyuan Xu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
50
|
Hu Y, Hu Q, Li Y, Lu L, Xiang Z, Yin Z, Kabelitz D, Wu Y. γδ T cells: origin and fate, subsets, diseases and immunotherapy. Signal Transduct Target Ther 2023; 8:434. [PMID: 37989744 PMCID: PMC10663641 DOI: 10.1038/s41392-023-01653-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/07/2023] [Accepted: 09/12/2023] [Indexed: 11/23/2023] Open
Abstract
The intricacy of diseases, shaped by intrinsic processes like immune system exhaustion and hyperactivation, highlights the potential of immune renormalization as a promising strategy in disease treatment. In recent years, our primary focus has centered on γδ T cell-based immunotherapy, particularly pioneering the use of allogeneic Vδ2+ γδ T cells for treating late-stage solid tumors and tuberculosis patients. However, we recognize untapped potential and optimization opportunities to fully harness γδ T cell effector functions in immunotherapy. This review aims to thoroughly examine γδ T cell immunology and its role in diseases. Initially, we elucidate functional differences between γδ T cells and their αβ T cell counterparts. We also provide an overview of major milestones in γδ T cell research since their discovery in 1984. Furthermore, we delve into the intricate biological processes governing their origin, development, fate decisions, and T cell receptor (TCR) rearrangement within the thymus. By examining the mechanisms underlying the anti-tumor functions of distinct γδ T cell subtypes based on γδTCR structure or cytokine release, we emphasize the importance of accurate subtyping in understanding γδ T cell function. We also explore the microenvironment-dependent functions of γδ T cell subsets, particularly in infectious diseases, autoimmune conditions, hematological malignancies, and solid tumors. Finally, we propose future strategies for utilizing allogeneic γδ T cells in tumor immunotherapy. Through this comprehensive review, we aim to provide readers with a holistic understanding of the molecular fundamentals and translational research frontiers of γδ T cells, ultimately contributing to further advancements in harnessing the therapeutic potential of γδ T cells.
Collapse
Affiliation(s)
- Yi Hu
- Microbiology and Immunology Department, School of Medicine, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Qinglin Hu
- Microbiology and Immunology Department, School of Medicine, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, 519000, China
| | - Yongsheng Li
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, 519000, China
| | - Zheng Xiang
- Microbiology and Immunology Department, School of Medicine, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Zhinan Yin
- Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong, 510632, China.
| | - Dieter Kabelitz
- Institute of Immunology, Christian-Albrechts-University Kiel, Kiel, Germany.
| | - Yangzhe Wu
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, 519000, China.
| |
Collapse
|