1
|
Peeters E, van Genugten EAJ, Heskamp S, de Vries IJM, van Herpen C, Koenen HJPM, Kneilling M, van der Post RS, van Dop WA, Westdorp H, Aarntzen E. Exploring molecular imaging to investigate immune checkpoint inhibitor-related toxicity. J Immunother Cancer 2025; 13:e011009. [PMID: 40341021 PMCID: PMC12060888 DOI: 10.1136/jitc-2024-011009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/18/2025] [Indexed: 05/10/2025] Open
Abstract
Immune checkpoint inhibitors (ICI) boost the endogenous anticancer immunity, evoking long-lasting anticancer responses in a subset of patients with solid tumors. Simultaneously, ICI are also associated with serious toxicities, impacting treatment duration and the quality of life. The proposed processes underlying ICI-related toxicity include T-cell activation and recruitment to non-tumor tissues, involvement of other immune cells and fibroblasts and the host' microbiome composition. However, the exact mechanisms of these processes remain incompletely understood, hindering clinicians' ability to predict and identify ICI-related toxicity in the early stages of treatment. Molecular imaging may play a role as a non-invasive biomarker, providing a tool to study ICI-related toxicity. This review discusses the applications of molecular imaging to answer questions regarding the mechanisms, detection, and prediction of ICI-related toxicity. Potential targets and the current state of development of suitable imaging techniques are discussed.
Collapse
Affiliation(s)
- Eva Peeters
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Sandra Heskamp
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carla van Herpen
- Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hans J P M Koenen
- Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, Gelderland, The Netherlands
| | - Manfred Kneilling
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, Tubingen, Baden-Württemberg, Germany
- Department of Dermatology, University of Tübingen, Tubingen, Baden-Württemberg, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls Universität Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Rachel S van der Post
- Department of Pathology, Radboud University Medical Center, Nijmegen, Gelderland, The Netherlands
| | - Willemijn A van Dop
- Department of Gastroenterology, Radboud University Medical Center, Nijmegen, Gelderland, The Netherlands
| | - Harm Westdorp
- Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Erik Aarntzen
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Nuclear Medicine, Eberhard Karls Universität Tübingen, Tübingen, Baden-Württemberg, Germany
- Department of Nuclear Medicine and Molecular Imaging, University Medical Centre Groningen, Groningen, Groningen, The Netherlands
| |
Collapse
|
2
|
Cao M, Deng Y, Hao Q, Yan H, Wang QL, Dong C, Wu J, He Y, Huang LB, Xia X, Gao Y, Chen HN, Zhang WH, Zhang YJ, Zhuo X, Dai L, Hu H, Peng Y, Zhang F, Liu Z, Huang W, Zhang H, Yang L, Shu Y, Zhang W, Zhang Y, Xu H. Single-cell transcriptomic analysis reveals gut microbiota-immunotherapy synergy through modulating tumor microenvironment. Signal Transduct Target Ther 2025; 10:140. [PMID: 40312419 PMCID: PMC12045981 DOI: 10.1038/s41392-025-02226-7] [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: 07/29/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 05/03/2025] Open
Abstract
The gut microbiota crucially regulates the efficacy of immune checkpoint inhibitor (ICI) based immunotherapy, but the underlying mechanisms remain unclear at the single-cell resolution. Using single-cell RNA sequencing and subsequent validations, we investigate gut microbiota-ICI synergy by profiling the tumor microenvironment (TME) and elucidating critical cellular interactions in mouse models. Our findings reveal that intact gut microbiota combined with ICIs may synergistically increase the proportions of CD8+, CD4+, and γδ T cells, reduce glycolysis metabolism, and reverse exhausted CD8+ T cells into memory/effector CD8+ T cells, enhancing antitumor response. This synergistic effect also induces macrophage reprogramming from M2 protumor Spp1+ tumor-associated macrophages (TAMs) to Cd74+ TAMs, which act as antigen-presenting cells (APCs). These macrophage subtypes show a negative correlation within tumors, particularly during fecal microbiota transplantation. Depleting Spp1+ TAMs in Spp1 conditional knockout mice boosts ICI efficacy and T cell infiltration, regardless of gut microbiota status, suggesting a potential upstream role of the gut microbiota and highlighting the crucial negative impact of Spp1+ TAMs during macrophage reprogramming on immunotherapy outcomes. Mechanistically, we propose a γδ T cell-APC-CD8+ T cell axis, where gut microbiota and ICIs enhance Cd40lg expression on γδ T cells, activating Cd40 overexpressing APCs (e.g., Cd74+ TAMs) through CD40-CD40L-related NF-κB signaling and boosting CD8+ T cell responses via CD86-CD28 interactions. These findings highlight the potential importance of γδ T cells and SPP1-related macrophage reprogramming in activating CD8+ T cells, as well as the synergistic effect of gut microbiota and ICIs in immunotherapy through modulating the TME.
Collapse
Affiliation(s)
- Minyuan Cao
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yun Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Hao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Huayun Yan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Quan-Lin Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China
| | - Chunyan Dong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yajiao He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Li-Bin Huang
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xuyang Xia
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yongchao Gao
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China
| | - Hai-Ning Chen
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Wei-Han Zhang
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yan-Jing Zhang
- Core Facilities, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaozhen Zhuo
- Department of Cardiology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Lunzhi Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hongbo Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Feng Zhang
- Center for Precision Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Zhaoqian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China
| | - Weihua Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China
| | - Huiyuan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Shu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China.
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
| | - Yan Zhang
- Lung Cancer Center/Lung Cancer Institute, Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, China.
| | - Heng Xu
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
- Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, China.
- Tianfu Jincheng Laboratory, Chengdu, China.
| |
Collapse
|
3
|
Nour MA, Rajabivahid M, Mehdi MSS, Tahmasebi S, Dashtgol SN, Dehghani-Ghorbi M, Vanan AG, Ghorbaninezhad F. A new era in melanoma immunotherapy: focus on DCs metabolic reprogramming. Cancer Cell Int 2025; 25:149. [PMID: 40234886 PMCID: PMC12001691 DOI: 10.1186/s12935-025-03781-3] [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: 12/11/2024] [Accepted: 04/03/2025] [Indexed: 04/17/2025] Open
Abstract
Melanoma, being one of the most dangerous forms of skin cancer, is characterized by its aggressive and metastatic nature, with the potential to develop resistance to various treatments. This resistance makes the disease challenging to treat, emphasizing the need for new treatment strategies. Within the tumor microenvironment (TME), melanoma cells exploit metabolic shifts, particularly glycolysis, to create an immunosuppressive TME that prevents dendritic cells (DCs) from functioning properly. Essential metabolic alterations such as lactate and lipid accumulation, and lack of tryptophan disrupt DC maturation, antigen presentation, and T cell activation. In recent years, melanoma immunotherapy has increasingly focused on reprogramming the metabolism of DCs. This review paper aims to provide insights into the metabolic suppression of melanoma-associated DCs, allowing the design of therapeutic strategies based on metabolic interventions to promote or restore DC function. This contribution reviews the metabolic reprogramming of DCs as a new approach for melanoma immunotherapy.
Collapse
Affiliation(s)
- Mina Afrashteh Nour
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mansour Rajabivahid
- Department of Internal Medicine, Valiasr Hospital, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Marjan Sadat Seyed Mehdi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Safa Tahmasebi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepideh Nasirzadeh Dashtgol
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Tehran Medical Branch, Islamic Azad University, Tehran, Iran
| | - Mahmoud Dehghani-Ghorbi
- Hematology-Oncology Department, Imam Hossein Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ahmad Ghorbani Vanan
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Farid Ghorbaninezhad
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Zitvogel L, Derosa L, Routy B, Loibl S, Heinzerling L, de Vries IJM, Engstrand L, Segata N, Kroemer G. Impact of the ONCOBIOME network in cancer microbiome research. Nat Med 2025; 31:1085-1098. [PMID: 40217075 DOI: 10.1038/s41591-025-03608-8] [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: 08/02/2024] [Accepted: 02/24/2025] [Indexed: 04/18/2025]
Abstract
The European Union-sponsored ONCOBIOME network has spurred an international effort to identify and validate relevant gut microbiota-related biomarkers in oncology, generating a unique and publicly available microbiome resource. ONCOBIOME explores the effects of the microbiota on gut permeability and metabolism as well as on antimicrobial and antitumor immune responses. Methods for the diagnosis of gut dysbiosis have been developed based on oncomicrobiome signatures associated with the diagnosis, prognosis and treatment responses in patients with cancer. The mechanisms explaining how dysbiosis compromises natural or therapy-induced immunosurveillance have been explored. Through its integrative approach of leveraging multiple cohorts across populations, cancer types and stages, ONCOBIOME has laid the theoretical and practical foundations for the recognition of microbiota alterations as a hallmark of cancer. ONCOBIOME has launched microbiota-centered interventions and lobbies in favor of official guidelines for avoiding diet-induced or iatrogenic (for example, antibiotic- or proton pump inhibitor-induced) dysbiosis. Here, we review the key advances of the ONCOBIOME network and discuss the progress toward translating these into oncology clinical practice.
Collapse
Affiliation(s)
- Laurence Zitvogel
- INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France.
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France.
- Clinicobiome, Gustave Roussy, Villejuif, France.
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France.
| | - Lisa Derosa
- INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- Clinicobiome, Gustave Roussy, Villejuif, France
| | - Bertrand Routy
- University of Montreal Research Center (CR-CHUM), Montreal, Quebec, Canada
- Department of Hematology-Oncology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - Sibylle Loibl
- German Breast Group c/ GBG Forschungs GmbH, Neu-Isenburg, Goethe University, Frankfurt, Germany
| | - Lucie Heinzerling
- Department of Dermatology and Allergy, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - I Jolanda M de Vries
- Medical Biosciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Lars Engstrand
- Department of Microbiology Tumor and Cell Biology, Centre for Translational Microbiome Research, Karolinska Institutet, Stockholm, Sweden
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- European Institute of Oncology IRCCS, Milan, Italy
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Institut Universitaire de France, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| |
Collapse
|
5
|
Alves Costa Silva C, Almonte AA, Zitvogel L. Oncobiomics: Leveraging Microbiome Translational Research in Immuno-Oncology for Clinical-Practice Changes. Biomolecules 2025; 15:504. [PMID: 40305219 PMCID: PMC12024955 DOI: 10.3390/biom15040504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/16/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Growing evidence suggests that cancer should not be viewed solely as a genetic disease but also as the result of functional defects in the metaorganism, including disturbances in the gut microbiota (i.e., gut dysbiosis). The human microbiota plays a critical role in regulating epithelial barrier function in the gut, airways, and skin, along with host metabolism and systemic immune responses against microbes and cancer. Collaborative international networks, such as ONCOBIOME, are essential in advancing research equity and building microbiome resources to identify and validate microbiota-related biomarkers and therapies. In this review, we explore the intricate relationship between the microbiome, metabolism, and cancer immunity, and we propose microbiota-based strategies to improve outcomes for individuals at risk of developing cancer or living with the disease.
Collapse
Affiliation(s)
- Carolina Alves Costa Silva
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif, France; (C.A.C.S.); (A.A.A.)
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée—Ligue Nationale Contre le Cancer, 94800 Villejuif, France
| | - Andrew A. Almonte
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif, France; (C.A.C.S.); (A.A.A.)
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée—Ligue Nationale Contre le Cancer, 94800 Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif, France; (C.A.C.S.); (A.A.A.)
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée—Ligue Nationale Contre le Cancer, 94800 Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, 94270 Kremlin-Bicêtre, France
- Center of Clinical Investigations BIOTHERIS, INSERM CIC1428, 94805 Villejuif, France
| |
Collapse
|
6
|
Motiño O, Lambertucci F, Joseph A, Durand S, Anagnostopoulos G, Li S, Carbonnier V, Nogueira-Recalde U, Montégut L, Chen H, Aprahamian F, Nirmalathasan N, Maiuri MC, Pietrocola F, Valla D, Laouénan C, Gautier JF, Castera L, Martins I, Kroemer G. ACBP/DBI neutralization for the experimental treatment of fatty liver disease. Cell Death Differ 2025; 32:434-446. [PMID: 39550516 PMCID: PMC11894144 DOI: 10.1038/s41418-024-01410-6] [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: 07/02/2024] [Revised: 10/09/2024] [Accepted: 10/28/2024] [Indexed: 11/18/2024] Open
Abstract
Acyl-CoA binding protein (ACBP), also known as diazepam-binding inhibitor (DBI), is an extracellular checkpoint of autophagy. Here, we report that patients with histologically confirmed metabolic-associated steatohepatitis (MASH) or liver fibrosis exhibit elevated levels of circulating ACBP/DBI protein as compared to non-affected controls. Plasma ACBP/DBI strongly correlated with the NAFLD and FIB4 scores in patients, and these correlations were independent of age and body mass index. We studied the capacity of a monoclonal antibody (mAb) neutralizing mouse ACBP/DBI to combat active liver disease in several mouse models, in which steatohepatitis had been induced by four different protocols, namely, (i) methionine/choline-deficient diet, (ii) Western style diet (WD) alone, (iii) WD combined with the hepatotoxic agent CCl4, and (iv) a combination of CCl4 injections and oral ethanol challenge. Injections of anti-ACBP/DBI mAb attenuated histological, enzymological, metabolomic and transcriptomic signs of liver damage in these four models, hence halting or reducing the progression of non-alcoholic and alcoholic liver disease. Steatosis, inflammation, ballooning and fibrosis responded to ACBP/DBI inhibition at the preclinical level. Altogether, these findings support a causal role of ACBP/DBI in MASH and liver fibrosis, as well as the possibility to therapeutically target ACBP/DBI.
Collapse
Grants
- O.M is supported by the “Beatriz Galindo Junior” Program from the Spanish Ministry of Universities (BG22/00104).
- UN-R is supported by “Axudas de apoio á etapa de formación posdoutoral da Xunta de Galicia” – GAIN. N°Expediente: IN606B-2021/015.
- DL, CL, J-FG and LC were supported by RHU QUID‐NASH funded by Agence Nationale de la Recherche programe Investissements d'Avenir (Reference ANR- 17-T171105J-RHUS-0009) implemented by Institut National de la Recherche Medicale, Université Paris Cité, Centre de l'Energie Atomique, Labortaoires Servier, Biopredictive, and Assistance Publique-Hôpitaux de Paris.
- GK is supported by the Ligue contre le Cancer (équipe labellisée); Agence National de la Recherche (ANR) – Projets blancs; Association pour la recherche sur le cancer (ARC); Cancéropôle Ile-de-France; Fondation pour la Recherche Médicale (FRM); a donation by Elior; European Joint Programme on Rare Diseases (EJPRD) Wilsonmed; European Research Council Advanced Investigator Award (ERC-2021-ADG, Grant No. 101052444; project acronym: ICD-Cancer, project title: Immunogenic cell death (ICD) in the cancer-immune dialogue), The ERA4 Health Cardinoff Grant Ener-LIGHT, European Union Horizon 2020 research and innovation programmes Oncobiome (grant agreement number: 825410, Project Acronym: ONCOBIOME, Project title: Gut OncoMicrobiome Signatures [GOMS] associated with cancer incidence, prognosis and prediction of treatment response, Prevalung (grant agreement number 101095604, Project Acronym: PREVALUNG EU, project title: Biomarkers affecting the transition from cardiovascular disease to lung cancer: towards stratified interception), Neutrocure (grant agreement number 861878 : Project Acronym: Neutrocure ; project title: Development of “smart” amplifiers of reactive oxygen species specific to aberrant polymorphonuclear neutrophils for treatment of inflammatory and autoimmune diseases, cancer and myeloablation); National support managed by the Agence Nationale de la Recherche under the France 2030 programme (reference number 21-ESRE-0028, ESR/Equipex+ Onco-Pheno-Screen); Hevolution Network on Senescence in Aging; 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; PAIR-Obésité INCa_1873, the RHUs Immunolife and LUCA-pi (both dedicated to France Relance 2030); Seerave Foundation; SIRIC Cancer Research and Personalized Medicine (CARPEM). This study contributes to the IdEx Université de Paris Cité ANR-18-IDEX-0001.
Collapse
Affiliation(s)
- Omar Motiño
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Unidad de Excelencia, Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid - CSIC, Valladolid, Spain.
| | - Flavia Lambertucci
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Adrien Joseph
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Sylvère Durand
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Gerasimos Anagnostopoulos
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Sijing Li
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Vincent Carbonnier
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Uxía Nogueira-Recalde
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Profesor Novoa Santos, A Coruña, Spain
| | - Léa Montégut
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Hui Chen
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Fanny Aprahamian
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Nitharsshini Nirmalathasan
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Maria Chiara Maiuri
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Department of Molecular Medicine and Medical Biotechnologies, University of Napoli Federico II, Naples, Italy
| | - Federico Pietrocola
- Department of Bioscience and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Dominique Valla
- Université Paris Cité, UMR1149 (CRI), Inserm, Paris, France
- Service hépatologie, AP-HP, Hôpital Beaujon, Clichy, France
| | - Cédric Laouénan
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm U1137, Laboratory "Infection, Antimicrobials, Modelling, Evolution" (IAME), Paris, France
- Département d'Epidémiologie Biostatistique et Recherche Clinique, AP-HP.Nord, Hôpital Bichat, Paris, France
| | - Jean-François Gautier
- Institut Necker Enfants Malades, Inserm U1151, CNRS UMR 8253, IMMEDIAB Laboratory, Paris, France
- Centre Universitaire de Diabétologie et de ses Complications, AP-HP, Hôpital Lariboisiére, Paris, France
| | - Laurent Castera
- Université Paris Cité, UMR1149 (CRI), Inserm, Paris, France
- Service hépatologie, AP-HP, Hôpital Beaujon, Clichy, France
| | - Isabelle Martins
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Institut Universitaire de France, Paris, France.
| |
Collapse
|
7
|
Pribyl AL, Hugenholtz P, Cooper MA. A decade of advances in human gut microbiome-derived biotherapeutics. Nat Microbiol 2025; 10:301-312. [PMID: 39779879 DOI: 10.1038/s41564-024-01896-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025]
Abstract
Microbiome science has evolved rapidly in the past decade, with high-profile publications suggesting that the gut microbiome is a causal determinant of human health. This has led to the emergence of microbiome-focused biotechnology companies and pharmaceutical company investment in the research and development of gut-derived therapeutics. Despite the early promise of this field, the first generation of microbiome-derived therapeutics (faecal microbiota products) have only recently been approved for clinical use. Next-generation therapies based on readily culturable and as-yet-unculturable colonic bacterial species (with the latter estimated to comprise 63% of all detected species) have not yet progressed to pivotal phase 3 trials. This reflects the many challenges involved in developing a new class of drugs in an evolving field. Here we discuss the evolution of the live biotherapeutics field over the past decade, from the development of first-generation products to the emergence of rationally designed second- and third-generation live biotherapeutics. Finally, we present our outlook for the future of this field.
Collapse
Affiliation(s)
| | - Philip Hugenholtz
- The University of Queensland, School of Chemistry and Molecular Biosciences, Australian Centre for Ecogenomics, Brisbane, Queensland, Australia.
| | | |
Collapse
|
8
|
Silva CAC, Fidelle M, Almonte AA, Derosa L, Zitvogel L. Gut Microbiota-Related Biomarkers in Immuno-Oncology. Annu Rev Pharmacol Toxicol 2025; 65:333-354. [PMID: 39259979 DOI: 10.1146/annurev-pharmtox-061124-102218] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Carcinogenesis is associated with the emergence of protracted intestinal dysbiosis and metabolic changes. Increasing evidence shows that gut microbiota-related biomarkers and microbiota-centered interventions are promising strategies to overcome resistance to immunotherapy. However, current standard methods for evaluating gut microbiota composition are cost- and time-consuming. The development of routine diagnostic tools for intestinal barrier alterations and dysbiosis constitutes a critical unmet medical need that can guide routine treatment and microbiota-centered intervention decisions in patients with cancer. In this review, we explore the influence of gut microbiota on cancer immunotherapy and highlight gut-associated biomarkers that have the potential to be transformed into simple diagnostic tools, thus guiding standard treatment decisions in the field of immuno-oncology. Mechanistic insights toward leveraging the complex relationship between cancer immunosurveillance, gut microbiota, and metabolism open exciting opportunities for developing novel biomarkers in immuno-oncology.
Collapse
Affiliation(s)
- Carolina Alves Costa Silva
- Clinicobiome, Gustave Roussy Cancer Campus (GRCC), and INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; ,
| | - Marine Fidelle
- Clinicobiome, Gustave Roussy Cancer Campus (GRCC), and INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; ,
| | - Andrew A Almonte
- Clinicobiome, Gustave Roussy Cancer Campus (GRCC), and INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; ,
| | - Lisa Derosa
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
- Clinicobiome, Gustave Roussy Cancer Campus (GRCC), and INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; ,
| | - Laurence Zitvogel
- Center of Clinical Investigations BIOTHERIS, INSERM CIC1428, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
- Clinicobiome, Gustave Roussy Cancer Campus (GRCC), and INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; ,
| |
Collapse
|
9
|
Khan M, Dong Y, Ullah R, Li M, Huang Q, Hu Y, Yang L, Luo Z. Recent Advances in Bacterium-Based Therapeutic Modalities for Melanoma Treatment. Adv Healthc Mater 2024; 13:e2401076. [PMID: 39375965 DOI: 10.1002/adhm.202401076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/16/2024] [Indexed: 10/09/2024]
Abstract
Melanoma is one of the most severe skin cancer indications with rapid progression and a high risk of metastasis. However, despite the accumulated advances in melanoma treatment including adjuvant radiation, chemotherapy, and immunotherapy, the overall melanoma treatment efficacy in the clinics is still not satisfactory. Interestingly, bacterial therapeutics have demonstrated unique properties for tumor-related therapeutic applications, such as tumor-targeted motility, tailorable cytotoxicity, and immunomodulatory capacity of the tumor microenvironment, which have emerged as a promising platform for melanoma therapy. Indeed, the recent advances in genetic engineering and nanotechnologies have boosted the application potential of bacterium-based therapeutics for treating melanoma by further enhancing their tumor-homing, cell-killing, drug delivery, and immunostimulatory capacities. This review provides a comprehensive summary of the state-of-the-art bacterium-based anti-melanoma modalities, which are categorized according to their unique functional merits, including tumor-specific cytotoxins, tumor-targeted drug delivery platforms, and immune-stimulatory agents. Furthermore, a perspective is provided discussing the potential challenges and breakthroughs in this area. The insights in this review may facilitate the development of more advanced bacterium-based therapeutic modalities for improved melanoma treatment efficacy.
Collapse
Affiliation(s)
- Mubassir Khan
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Yilong Dong
- Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325016, P. R. China
| | - Razi Ullah
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Lab for Vascular Implants College of Bioengineering Chongqing University, Chongqing, 400030, P. R. China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing, 400044, P. R. China
| | - Qiping Huang
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing, 400044, P. R. China
| |
Collapse
|
10
|
Hossain SM, Ly K, Sung YJ, Braithwaite A, Li K. Immune Checkpoint Inhibitor Therapy for Metastatic Melanoma: What Should We Focus on to Improve the Clinical Outcomes? Int J Mol Sci 2024; 25:10120. [PMID: 39337605 PMCID: PMC11432671 DOI: 10.3390/ijms251810120] [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: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have transformed cancer treatment by enhancing anti-tumour immune responses, demonstrating significant efficacy in various malignancies, including melanoma. However, over 50% of patients experience limited or no response to ICI therapy. Resistance to ICIs is influenced by a complex interplay of tumour intrinsic and extrinsic factors. This review summarizes current ICIs for melanoma and the factors involved in resistance to the treatment. We also discuss emerging evidence that the microbiota can impact ICI treatment outcomes by modulating tumour biology and anti-tumour immune function. Furthermore, microbiota profiles may offer a non-invasive method for predicting ICI response. Therefore, future research into microbiota manipulation could provide cost-effective strategies to enhance ICI efficacy and improve outcomes for melanoma patients.
Collapse
Affiliation(s)
- Sultana Mehbuba Hossain
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Kevin Ly
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Yih Jian Sung
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Antony Braithwaite
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Kunyu Li
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| |
Collapse
|
11
|
Chen H, Moriceau S, Joseph A, Mailliet F, Li S, Tolle V, Duriez P, Dardennes R, Durand S, Carbonnier V, Stoll G, Sauvat A, Lachkar S, Aprahamian F, Alves Costa Silva C, Pan H, Montégut L, Anagnostopoulos G, Lambertucci F, Motiño O, Nogueira-Recalde U, Bourgin M, Mao M, Pan Y, Cerone A, Boedec E, Gouveia ZL, Marmorino F, Cremolini C, Derosa L, Zitvogel L, Kepp O, López-Otín C, Maiuri MC, Perez F, Gorwood P, Ramoz N, Oury F, Martins I, Kroemer G. Acyl-CoA binding protein for the experimental treatment of anorexia. Sci Transl Med 2024; 16:eadl0715. [PMID: 39141698 DOI: 10.1126/scitranslmed.adl0715] [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: 09/27/2023] [Revised: 03/25/2024] [Accepted: 07/17/2024] [Indexed: 08/16/2024]
Abstract
Extracellular acyl-coenzyme A binding protein [ACBP encoded by diazepam binding inhibitor (DBI)] is a phylogenetically ancient appetite stimulator that is secreted in a nonconventional, autophagy-dependent fashion. Here, we show that low ACBP/DBI plasma concentrations are associated with poor prognosis in patients with anorexia nervosa, a frequent and often intractable eating disorder. In mice, anorexia induced by chronic restraint stress (CRS) is accompanied by a reduction in circulating ACBP/DBI concentrations. We engineered a chemical-genetic system for the secretion of ACBP/DBI through a biotin-activatable, autophagy-independent pathway. In transgenic mice expressing this system in hepatocytes, biotin-induced elevations in plasma ACBP/DBI concentrations prevented anorexia induced by CRS or chemotherapeutic agents including cisplatin, doxorubicin, and paclitaxel. ACBP/DBI reversed the CRS or cisplatin-induced increase in plasma lipocalin-2 concentrations and the hypothalamic activation of anorexigenic melanocortin 4 receptors, for which lipocalin-2 is an agonist. Daily intravenous injections of recombinant ACBP/DBI protein or subcutaneous implantation of osmotic pumps releasing recombinant ACBP/DBI mimicked the orexigenic effects of the chemical-genetic system. In conclusion, the supplementation of extracellular and peripheral ACBP/DBI might constitute a viable strategy for treating anorexia.
Collapse
Affiliation(s)
- Hui Chen
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, 91400 Paris, France
| | - Stéphanie Moriceau
- Institut Imagine, Platform for Neurobehavioral and Metabolism, Structure Fédérative de Recherche Necker, 26 INSERM US24/CNRS UAR, 3633, 75015 Paris, France
| | - Adrien Joseph
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Service de Réanimation Médicale, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris, 75010 Paris, France
| | - Francois Mailliet
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 8, F-75015 Paris, France
| | - Sijing Li
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, 91400 Paris, France
| | - Virginie Tolle
- Université de Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Genetic Vulnerability to Addictive and Psychiatric Disorders Team, 75015 Paris, France
| | - Philibert Duriez
- Université de Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Genetic Vulnerability to Addictive and Psychiatric Disorders Team, 75015 Paris, France
- Université Paris Cité and GHU Paris Psychiatrie et Neurosciences, CMME, Hôpital Sainte-Anne, 75014 Paris, France
| | - Roland Dardennes
- Université Paris Cité and GHU Paris Psychiatrie et Neurosciences, CMME, Hôpital Sainte-Anne, 75014 Paris, France
| | - Sylvère Durand
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
| | - Vincent Carbonnier
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
| | - Gautier Stoll
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
| | - Allan Sauvat
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
| | - Sylvie Lachkar
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
| | - Fanny Aprahamian
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
| | - Carolina Alves Costa Silva
- Gustave Roussy Cancer Campus, 94805 Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, 94800 Le Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée-Ligue Nationale contre le Cancer, 94805 Villejuif, France
| | - Hui Pan
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, 91400 Paris, France
| | - Léa Montégut
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, 91400 Paris, France
| | - Gerasimos Anagnostopoulos
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
| | - Flavia Lambertucci
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
| | - Omar Motiño
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
| | - Uxía Nogueira-Recalde
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
- Rheumatology Research Group (GIR), Biomedical Research Institute of A Coruña (INIBIC), Professor Novoa Santos Foundation, 15006 A Coruña, Spain
| | - Mélanie Bourgin
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
| | - Misha Mao
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, 91400 Paris, France
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, 310016 Hangzhou, Zhejiang, China
| | - Yuhong Pan
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, 91400 Paris, France
| | - Alexandra Cerone
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
| | - Erwan Boedec
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Biochemistry and Biophysics (B&B) Core Facility, 75014 Paris, France
| | - Zelia L Gouveia
- Cell Biology and Cancer Unit, Institut Curie, PSL Research University, CNRS, 75005 Paris, France
| | - Federica Marmorino
- Unit of Medical Oncology 2, Azienda Ospedaliero Universitaria Pisana, 56126 Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Chiara Cremolini
- Unit of Medical Oncology 2, Azienda Ospedaliero Universitaria Pisana, 56126 Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Lisa Derosa
- Gustave Roussy Cancer Campus, 94805 Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, 94800 Le Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée-Ligue Nationale contre le Cancer, 94805 Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, 94805 Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, 94800 Le Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée-Ligue Nationale contre le Cancer, 94805 Villejuif, France
| | - Oliver Kepp
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
| | - Carlos López-Otín
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Facultad de Ciencias de la Vida y la Naturaleza, Universidad Nebrija, 28248 Madrid, Spain
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain
| | - Maria Chiara Maiuri
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Naples, Italy
| | - Franck Perez
- Cell Biology and Cancer Unit, Institut Curie, PSL Research University, CNRS, 75005 Paris, France
| | - Philip Gorwood
- Université de Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Genetic Vulnerability to Addictive and Psychiatric Disorders Team, 75015 Paris, France
- Université Paris Cité and GHU Paris Psychiatrie et Neurosciences, CMME, Hôpital Sainte-Anne, 75014 Paris, France
| | - Nicolas Ramoz
- Université de Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Genetic Vulnerability to Addictive and Psychiatric Disorders Team, 75015 Paris, France
- Université Paris Cité and GHU Paris Psychiatrie et Neurosciences, CMME, Hôpital Sainte-Anne, 75014 Paris, France
| | - Franck Oury
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 8, F-75015 Paris, France
| | - Isabelle Martins
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, 94805 Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France
| |
Collapse
|
12
|
Bredon M, Danne C, Pham HP, Ruffié P, Bessede A, Rolhion N, Creusot L, Brot L, Alonso I, Langella P, Derosa L, Cortot AB, Routy B, Zitvogel L, Segata N, Sokol H. Faecalibaterium prausnitzii strain EXL01 boosts efficacy of immune checkpoint inhibitors. Oncoimmunology 2024; 13:2374954. [PMID: 38957477 PMCID: PMC11218805 DOI: 10.1080/2162402x.2024.2374954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024] Open
Abstract
Gut microbiota impacts responses to immune checkpoint inhibitors (ICI). A high level of Faecalibacterium prausnitzii have been associated with a positive response to ICI in multiple cancer types. Here, based on fecal shotgun metagenomics data, we show in two independent cohorts of patients with non-small cell lung cancer and advanced melanoma that a high level of F. prausnitzii at baseline is positively associated with a better clinical response to ICI. In MCA205 tumor-bearing mice, administration of F. prausnitzii strain EXL01, already in clinical development for Inflammatory Bowel Disease, restores the anti-tumor response to ICI in the context of antibiotic-induced microbiota perturbation at clinical and tumor transcriptomics level. In vitro, EXL01 strain enhances T cell activation in the presence of ICI. Interestingly, oral administration of EXL01 strain did not induce any change in fecal microbiota diversity or composition, suggesting a direct effect on immune cells in the small intestine. F. prausnitzii strain EXL01 will be evaluated as an adjuvant to ICI in multiple cancers in the near future.
Collapse
Affiliation(s)
- Marius Bredon
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Camille Danne
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | | | | | | | - Nathalie Rolhion
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Laura Creusot
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Loic Brot
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Iria Alonso
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Philippe Langella
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, Jouy-en-Josas, France
| | - Lisa Derosa
- UMR1015 Gustave Roussy Cancer Campus, Villejuif, France
- Département de Médecine Oncologique, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicetre, France
| | - Alexis B. Cortot
- Université de Lille, CHU Lille, Thoracic Oncology Department, Centre National de la Recherche Scientifique, INSERM, Institut Pasteur de Lille, UMR9020-UMR-S 1277-Canther, Lille, France
| | - Bertrand Routy
- Department of Medicine, Hematology-Oncology Division, University of Montreal Healthcare Centre (CHUM), Montreal, QC, Canada
- University of Montreal Research Center (CRCHUM), Montreal, QC, Canada
| | - Laurence Zitvogel
- UMR1015 Gustave Roussy Cancer Campus, Villejuif, France
- Département de Médecine Oncologique, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicetre, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Harry Sokol
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, Jouy-en-Josas, France
| |
Collapse
|
13
|
Bol KF, Schreibelt G, Bloemendal M, van Willigen WW, Hins-de Bree S, de Goede AL, de Boer AJ, Bos KJH, Duiveman-de Boer T, Olde Nordkamp MAM, van Oorschot TGM, Popelier CJ, Pots JM, Scharenborg NM, van de Rakt MWMM, de Ruiter V, van Meeteren WS, van Rossum MM, Croockewit SJ, Koeneman BJ, Creemers JHA, Wortel IMN, Angerer C, Brüning M, Petry K, Dzionek A, van der Veldt AA, van Grünhagen DJ, Werner JEM, Bonenkamp JJ, Haanen JBAG, Boers-Sonderen MJ, Koornstra RHT, Boomsma MF, Aarntzen EHJ, Gotthardt M, Nagarajah J, de Witte TJM, Figdor CG, de Wilt JHW, Textor J, de Groot JWB, Gerritsen WR, de Vries IJM. Adjuvant dendritic cell therapy in stage IIIB/C melanoma: the MIND-DC randomized phase III trial. Nat Commun 2024; 15:1632. [PMID: 38395969 PMCID: PMC10891118 DOI: 10.1038/s41467-024-45358-0] [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: 05/09/2023] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Autologous natural dendritic cells (nDCs) treatment can induce tumor-specific immune responses and clinical responses in cancer patients. In this phase III clinical trial (NCT02993315), 148 patients with resected stage IIIB/C melanoma were randomized to adjuvant treatment with nDCs (n = 99) or placebo (n = 49). Active treatment consisted of intranodally injected autologous CD1c+ conventional and plasmacytoid DCs loaded with tumor antigens. The primary endpoint was the 2-year recurrence-free survival (RFS) rate, whereas the secondary endpoints included median RFS, 2-year and median overall survival, adverse event profile, and immunological response The 2-year RFS rate was 36.8% in the nDC treatment group and 46.9% in the control group (p = 0.31). Median RFS was 12.7 months vs 19.9 months, respectively (hazard ratio 1.25; 90% CI: 0.88-1.79; p = 0.29). Median overall survival was not reached in both treatment groups (hazard ratio 1.32; 90% CI: 0.73-2.38; p = 0.44). Grade 3-4 study-related adverse events occurred in 5% and 6% of patients. Functional antigen-specific T cell responses could be detected in 67.1% of patients tested in the nDC treatment group vs 3.8% of patients tested in the control group (p < 0.001). In conclusion, while adjuvant nDC treatment in stage IIIB/C melanoma patients generated specific immune responses and was well tolerated, no benefit in RFS was observed.
Collapse
Affiliation(s)
- 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
| | - Gerty Schreibelt
- Medical Biosciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Martine Bloemendal
- 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
| | - Wouter W van Willigen
- 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
| | - Simone Hins-de Bree
- Medical Biosciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Anna L de Goede
- Department of Pharmacy, Radboud university medical center, Nijmegen, The Netherlands
| | - Annemiek J de Boer
- Medical Biosciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Kevin J H Bos
- Medical Biosciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Tjitske Duiveman-de Boer
- Medical Biosciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Michel A M Olde Nordkamp
- Medical Biosciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Tom G M van Oorschot
- Medical Biosciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Carlijn J Popelier
- Medical Biosciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Jeanne M Pots
- Medical Biosciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Nicole M Scharenborg
- Medical Biosciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Mandy W M M van de Rakt
- Medical Biosciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Valeska de Ruiter
- Medical Biosciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Wilmy S van Meeteren
- Department of Dermatology, Radboud university medical center, Nijmegen, The Netherlands
| | - Michelle M van Rossum
- Department of Dermatology, Radboud university medical center, Nijmegen, The Netherlands
| | - Sandra J Croockewit
- Department of Hematology, Radboud university medical center, Nijmegen, The Netherlands
| | - Bouke J Koeneman
- Medical Biosciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Jeroen H A Creemers
- Medical Biosciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Inge M N Wortel
- Medical Biosciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
- Department of Data Science, Institute for Computing and Information Sciences, Radboud University, Nijmegen, The Netherlands
| | | | | | | | | | - Astrid A van der Veldt
- Departments of Medical Oncology and Radiology & Nuclear Medicine, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Dirk J van Grünhagen
- Department Surgical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Johanna E M Werner
- Department Surgical Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | - Johannes J Bonenkamp
- Department Surgical Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | - John B A G Haanen
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marye J Boers-Sonderen
- Department of Medical Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | - Rutger H T Koornstra
- Department of Medical Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | - Martijn F Boomsma
- Department of Radiology, Isala Oncology Center, Zwolle, The Netherlands
| | - Erik H J Aarntzen
- Department of Medical Imaging, Radboud university medical center, Nijmegen, The Netherlands
| | - Martin Gotthardt
- Department of Medical Imaging, Radboud university medical center, Nijmegen, The Netherlands
| | - James Nagarajah
- Department of Medical Imaging, Radboud university medical center, Nijmegen, The Netherlands
| | - Theo J M de Witte
- Medical Biosciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Carl G Figdor
- Medical Biosciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Johannes H W de Wilt
- Department Surgical Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | - Johannes Textor
- Medical Biosciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
- Department of Data Science, Institute for Computing and Information Sciences, Radboud University, Nijmegen, The Netherlands
| | | | - Winald R Gerritsen
- 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
|