1
|
Talebi F, Gregucci F, Ahmed J, Ben Chetrit N, D. Brown B, Chan TA, Chand D, Constanzo J, Demaria S, I. Gabrilovich D, Golden E, Godkin A, Guha C, P. Gupta G, Hasan A, G. Herrera F, Kaufman H, Li D, A. Melcher A, McDonald S, Merghoub T, Monjazeb AM, Paris S, Pitroda S, Sadanandam A, Schaue D, Santambrogio L, Szapary P, Sage J, W. Welsh J, Wilkins A, H. Young K, Wennerberg E, Zitvogel L, Galluzzi L, Deutsch E, C. Formenti S. Updates on radiotherapy-immunotherapy combinations: Proceedings of 8th Annual ImmunoRad Conference. Oncoimmunology 2025; 14:2507856. [PMID: 40401900 PMCID: PMC12101595 DOI: 10.1080/2162402x.2025.2507856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/23/2025] Open
Abstract
The annual ImmunoRad Conference has established itself as a recurrent occasion to explore the possibility of combining radiation therapy (RT) and immunotherapy (IT) for clinical cancer management. Bringing together a number of preclinical and clinical leaders in the fields of radiation oncology, immuno-oncology and IT, this annual event fosters indeed essential conversations and fruitful exchanges on how to address existing challenges to expand the therapeutic value of RT-IT combinations. The 8th edition of the ImmunoRad Conference, which has been held in October 2024 at the Weill Cornell Medical College of New York City, highlighted exciting preclinical and clinical advances at the interface between RT and IT, setting the stage for extra progress toward extended benefits for patients with an increasing variety of tumor types. Here, we critically summarize the lines of investigation that have been discussed at the occasion of the 8th Annual ImmunoRad Conference.
Collapse
Affiliation(s)
- Fereshteh Talebi
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Fabiana Gregucci
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Jalal Ahmed
- Icahn Genomics Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nir Ben Chetrit
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Brian D. Brown
- Icahn Genomics Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Timothy A. Chan
- Department of Cancer Sciences, Global Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Case Western University School of Medicine, Cleveland, OH, USA
| | | | - Julie Constanzo
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | | | - Encouse Golden
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Andrew Godkin
- Division of Infection and Immunity/Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff, UK
| | - Chandan Guha
- Departments of Radiation Oncology and Pathology, Albert Einstein College of Medicine, New York, NY, USA
| | - Gaorav P. Gupta
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Fernanda G. Herrera
- AGORA Cancer Research Center, Swiss Cancer Center Leman, Lausanne, Switzerland
- Services of Radiation Oncology and Immuno-Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- Department of Oncology, Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland
| | | | - Donna Li
- University of Wisconsin, Madison, WI, USA
| | - Alan A. Melcher
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Sierra McDonald
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center and Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, NY, USA
| | - Arta M. Monjazeb
- Department of Radiation Oncology, University of California, San Diego, CA, USA
| | | | - Sean Pitroda
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Anguraj Sadanandam
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | | | - Julien Sage
- Departments of Genetics and Pediatrics, Stanford University, Stanford, California
| | - James W. Welsh
- Department of Radiation Oncology, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Anna Wilkins
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Kristina H. Young
- Division of Radiation Oncology, The Oregon Clinic, Portland, OR, USA
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Eric Wennerberg
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Laurence Zitvogel
- Gustave Roussy, INSERM U1015, Division of Medicine, Paris-Saclay University, Center of Clinical Investigations BIOTHERIS, Villejuif, France
| | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy, INSERM U1030, Division of Medicine, Paris-Saclay University, RHU LySAIRI “Lymphocyte-Sparing Artificial Intelligence-guided Radio-Immunotherapy”, Villejuif, France
| | - Silvia C. Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
2
|
Galluzzi L, Buqué A. Paradoxical control of multifocal mammary oncogenesis by radiation therapy. Oncoimmunology 2025; 14:2458886. [PMID: 39873285 PMCID: PMC11776481 DOI: 10.1080/2162402x.2025.2458886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/04/2025] [Accepted: 01/08/2025] [Indexed: 01/30/2025] Open
Abstract
In an immunocompetent mouse model of multifocal, metachronous HR+ mammary carcinogenesis, we have recently demonstrated that a superior control of primary neoplastic lesions by focal radiotherapy does not necessarily translate into improved oncosuppression at non-irradiated (pre)malignant tissues. These data point to a link between local tumor control by radiotherapy and systemic oncogenesis that remains to be fully understood.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Aitziber Buqué
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
3
|
Jaa A, Buque A. Novel metabolic routes to cancer immune evasion. Trends Cancer 2025:S2405-8033(25)00125-6. [PMID: 40393916 DOI: 10.1016/j.trecan.2025.05.002] [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: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/22/2025]
Abstract
The tumor microenvironment (TME) comprises heterogeneous cell types that closely interact with each other. Crosstalk among the TME components determines antitumor immune responses and their sensitivity to therapies such as immunotherapy. Recent studies published in Cancer Cell by Tang et al. and Zhu et al. identify two novel metabolic adaptations that tumors use to facilitate immune evasion. These targetable mechanisms suggest new avenues to improve antitumor immune responses.
Collapse
Affiliation(s)
- Ayoub Jaa
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Aitziber Buque
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Yan Y, Yu J, Wang W, Xu Y, Tison K, Xiao R, Grove S, Wei S, Vatan L, Wicha M, Kryczek I, Zou W. Palmitoylation prevents B7-H4 lysosomal degradation sustaining tumor immune evasion. Nat Commun 2025; 16:4254. [PMID: 40341398 PMCID: PMC12062253 DOI: 10.1038/s41467-025-58552-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 03/26/2025] [Indexed: 05/10/2025] Open
Abstract
B7-H4 functions as an immune checkpoint in the tumor microenvironment (TME). However, the post-translational modification (PTM) of B7-H4 and its translational potential in cancer remains incompletely understood. We find that ZDHHC3, a zinc finger DHHC-type palmitoyltransferase, palmitoylates B7-H4 at Cys130 in breast cancer cells, preventing its lysosomal degradation and sustaining B7-H4-mediated immunosuppression. Knockdown of ZDHHC3 in tumors results in robust anti-tumor immunity and reduces tumor progression in murine models. Moreover, abemaciclib, a CDK4/6 inhibitor, primes lysosome activation and promotes lysosomal degradation of B7-H4 independently of the tumor cell cycle. Treatment with abemaciclib results in T cell activation and mitigates B7-H4-mediated immune suppression via inducing B7-H4 degradation in preclinical tumor models. Thus, B7-H4 palmitoylation is an important PTM controlling B7-H4 protein stability and abemaciclib may be repurposed to promote B7-H4 degradation, thereby treating patients with B7-H4 expressing tumors.
Collapse
Affiliation(s)
- Yijian Yan
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Jiali Yu
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Weichao Wang
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Ying Xu
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Kole Tison
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Rongxin Xiao
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Sara Grove
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Shuang Wei
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Linda Vatan
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Max Wicha
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ilona Kryczek
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Weiping Zou
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA.
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA.
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA.
- Graduate Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
5
|
Buqué A, Bloy N, Petroni G, Jiménez-Cortegana C, Sato A, Iribarren C, Yamazaki T, Galassi C, Hensler M, Bhinder B, Guarracino A, Rippon B, Beltran-Visiedo M, Soler-Agesta R, Pannellini T, Fucikova J, Demaria S, Zhou XK, Elemento O, Formenti SC, Galluzzi L. Impact of radiation therapy dose, fractionation, and immunotherapeutic partner in a mouse model of hormone receptor-positive mammary carcinogenesis. J Natl Cancer Inst 2025; 117:934-947. [PMID: 39661487 PMCID: PMC12058254 DOI: 10.1093/jnci/djae329] [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: 10/18/2024] [Revised: 11/26/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Hormone receptor-positive (HR+) breast cancer responds poorly to immune checkpoint inhibitors (ICIs). In some settings, radiation therapy (RT) has been shown to mediate immunostimulatory effects and promote ICI sensitivity. METHODS We investigated whether hypofractionated RT may be successfully combined with ICIs in a mouse model of multifocal, metachronous HR+ mammary carcinogenesis. We hypothesized that focal RT targeting the first detectable (primary) tumor combined with ICIs may generate effective immunity, delaying the development of new lesions. RESULTS Focal RT in various doses and fractionations limited primary tumor growth, with an optimum for a 20-Gy × 2 regimen (ablative in approximately 90% of mice). The degree of primary disease control, however, did not necessarily correlate with overall survival extension because of changes in the development of new neoplastic lesions contributing to global tumor burden. Adding a PD-1 blocker to focal RT delivered in a 10-Gy × 3, 20-Gy × 2, or 8-Gy × 6 regimen failed to alter overall survival extension enabled by RT alone. Similar results were obtained with a CTLA4 blocker, an IL-1β inhibitor, and a PD-1 blocker plus recombinant FLT3LG when combined with the 10-Gy × 3 regimen. CONCLUSIONS In this model of HR+ mammary carcinogenesis, RT to the primary tumor ameliorates overall survival (to an extent based on dose and fractionation). Increasing local control through RT alone or RT plus immunotherapy beyond a hitherto undefined threshold, however, does not necessarily inhibit the development of subsequent nonirradiated neoplasms and hence does not necessarily provide extra overall survival benefits.
Collapse
Affiliation(s)
- Aitziber Buqué
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, United States
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadephia, PA 19111, United States
| | - Norma Bloy
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, United States
| | - Giulia Petroni
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, United States
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | | | - Ai Sato
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, United States
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadephia, PA 19111, United States
| | - Cristina Iribarren
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, United States
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, United States
| | - Claudia Galassi
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, United States
| | | | - Bhavneet Bhinder
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, United States
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY 10065, United States
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, United States
| | - Andrea Guarracino
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, United States
- Human Technopole, Milan 20157, Italy
| | - Brady Rippon
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065, United States
| | - Manuel Beltran-Visiedo
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, United States
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadephia, PA 19111, United States
| | - Ruth Soler-Agesta
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, United States
- Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza/Aragón Health Research Institute, Zaragoza 50009, Spain
| | - Tania Pannellini
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, United States
| | - Jitka Fucikova
- Sotio, Prague 170 00, Czech Republic
- Department of Immunology, Charles University, 2nd Faculty of Medicine, and University Hospital Motol, Prague 150 06, Czech Republic
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, United States
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, United States
| | - Xi Kathy Zhou
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065, United States
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY 10065, United States
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, United States
- Sandra and Edward Meyer Cancer Center, New York, NY 10065, United States
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, United States
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, United States
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadephia, PA 19111, United States
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY 10065, United States
- Sandra and Edward Meyer Cancer Center, New York, NY 10065, United States
| |
Collapse
|
6
|
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
|
7
|
Chen J, Levy A, Tian AL, Huang X, Cai G, Fidelle M, Rauber C, Ly P, Pizzato E, Sitterle L, Piccinno G, Liu P, Durand S, Mao M, Zhao L, Iebba V, Felchle H, de La Varende ALM, Fischer JC, Thomas S, Greten TF, Jones JC, Monge C, Demaria S, Formenti S, Belluomini L, Dionisi V, Massard C, Blanchard P, Robert C, Quevrin C, Lopes E, Clémenson C, Mondini M, Meziani L, Zhan Y, Zeng C, Cai Q, Morel D, Sun R, Laurent PA, Mangoni M, Di Cataldo V, Arilli C, Trommer M, Wegen S, Neppl S, Riechelmann RP, Camandaroba MP, Neto ES, Fournier PE, Segata N, Holicek P, Galluzzi L, Aitziber B, Silva CAC, Derosa L, Kroemer G, Chen C, Zitvogel L, Deutsch E. Low-dose irradiation of the gut improves the efficacy of PD-L1 blockade in metastatic cancer patients. Cancer Cell 2025; 43:361-379.e10. [PMID: 40068595 PMCID: PMC11907695 DOI: 10.1016/j.ccell.2025.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 11/03/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025]
Abstract
The mechanisms governing the abscopal effects of local radiotherapy in cancer patients remain an open conundrum. Here, we show that off-target intestinal low-dose irradiation (ILDR) increases the clinical benefits of immune checkpoint inhibitors or chemotherapy in eight retrospective cohorts of cancer patients and in tumor-bearing mice. The abscopal effects of ILDR depend on dosimetry (≥1 and ≤3 Gy) and on the metabolic and immune host-microbiota interaction at baseline allowing CD8+ T cell activation without exhaustion. Various strains of Christensenella minuta selectively boost the anti-cancer efficacy of ILDR and PD-L1 blockade, allowing emigration of intestinal PD-L1-expressing dendritic cells to tumor-draining lymph nodes. An interventional phase 2 study provides the proof-of-concept that ILDR can circumvent resistance to first- or second-line immunotherapy in cancer patients. Prospective clinical trials are warranted to define optimal dosimetry and indications for ILDR to maximize its therapeutic potential.
Collapse
Affiliation(s)
- Jianzhou Chen
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif Cedex, France
- Faculté de Médecine, Université Paris-Saclay, 94270 Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR 1015, Equipe Labellisée—Ligue Nationale contre le Cancer, 94805 Villejuif, France
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, China
| | - Antonin Levy
- Faculté de Médecine, Université Paris-Saclay, 94270 Kremlin-Bicêtre, France
- Department of Radiation Oncology, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
- INSERM U1030, Radiothérapie Moléculaire et Innovations Thérapeutiques, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
| | - Ai-Ling Tian
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif Cedex, France
- Faculté de Médecine, Université Paris-Saclay, 94270 Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR 1015, Equipe Labellisée—Ligue Nationale contre le Cancer, 94805 Villejuif, France
| | - Xuehan Huang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, China
| | - Guoxin Cai
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif Cedex, France
- Faculté de Médecine, Université Paris-Saclay, 94270 Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR 1015, Equipe Labellisée—Ligue Nationale contre le Cancer, 94805 Villejuif, France
| | - Marine Fidelle
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR 1015, Equipe Labellisée—Ligue Nationale contre le Cancer, 94805 Villejuif, France
- CICBT1428, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
| | - Conrad Rauber
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR 1015, Equipe Labellisée—Ligue Nationale contre le Cancer, 94805 Villejuif, France
- Department of Gastroenterology and Infectious Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Pierre Ly
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif Cedex, France
- Faculté de Médecine, Université Paris-Saclay, 94270 Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR 1015, Equipe Labellisée—Ligue Nationale contre le Cancer, 94805 Villejuif, France
| | - Eugénie Pizzato
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif Cedex, France
- Faculté de Médecine, Université Paris-Saclay, 94270 Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR 1015, Equipe Labellisée—Ligue Nationale contre le Cancer, 94805 Villejuif, France
| | - Lisa Sitterle
- INSERM U1030, Radiothérapie Moléculaire et Innovations Thérapeutiques, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
| | - Gianmarco Piccinno
- Department of Computational, Cellular and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Peng Liu
- Metabolomics and Cell Biology Platforms, UMS AMMICa, Gustave Roussy Cancer Campus, 94805 Villejuif, France
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée – Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, 75006 Paris, France
| | - Sylvère Durand
- Metabolomics and Cell Biology Platforms, UMS AMMICa, Gustave Roussy Cancer Campus, 94805 Villejuif, France
| | - Misha Mao
- Metabolomics and Cell Biology Platforms, UMS AMMICa, Gustave Roussy Cancer Campus, 94805 Villejuif, France
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée – Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310000, Zhejiang, China
| | - Liwei Zhao
- Metabolomics and Cell Biology Platforms, UMS AMMICa, Gustave Roussy Cancer Campus, 94805 Villejuif, France
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée – Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, 75006 Paris, France
| | - Valerio Iebba
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif Cedex, France
| | - Hannah Felchle
- Metabolomics and Cell Biology Platforms, UMS AMMICa, Gustave Roussy Cancer Campus, 94805 Villejuif, France
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée – Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Technical University of Munich (TUM), TUM School of Medicine and Health, Klinikum rechts der Isar, Department of Radiation Oncology, 81675 Munich, Germany
| | - Anne-Laure Mallard de La Varende
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif Cedex, France
- Faculté de Médecine, Université Paris-Saclay, 94270 Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR 1015, Equipe Labellisée—Ligue Nationale contre le Cancer, 94805 Villejuif, France
| | - Julius Clemens Fischer
- Metabolomics and Cell Biology Platforms, UMS AMMICa, Gustave Roussy Cancer Campus, 94805 Villejuif, France
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée – Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Technical University of Munich (TUM), TUM School of Medicine and Health, Klinikum rechts der Isar, Department of Radiation Oncology, 81675 Munich, Germany
| | - Simon Thomas
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif Cedex, France
- Faculté de Médecine, Université Paris-Saclay, 94270 Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR 1015, Equipe Labellisée—Ligue Nationale contre le Cancer, 94805 Villejuif, France
| | - Tim F. Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jennifer C. Jones
- Translational Nanobiology Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cecilia Monge
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Silvia Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lorenzo Belluomini
- Section of Innovation Biomedicine - Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, 37134 Verona, Italy
| | - Valeria Dionisi
- Department of Radiation Oncology, University of Verona Hospital Trust, 37126 Verona, Italy
| | - Christophe Massard
- Faculté de Médecine, Université Paris-Saclay, 94270 Kremlin-Bicêtre, France
- INSERM U1030, Radiothérapie Moléculaire et Innovations Thérapeutiques, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
- Drug Development Department (DITEP), Gustave Roussy-Cancer Campus, 94805 Villejuif, France
| | - Pierre Blanchard
- Faculté de Médecine, Université Paris-Saclay, 94270 Kremlin-Bicêtre, France
- INSERM U1030, Radiothérapie Moléculaire et Innovations Thérapeutiques, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
| | - Charlotte Robert
- INSERM U1030, Radiothérapie Moléculaire et Innovations Thérapeutiques, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
| | - Clément Quevrin
- INSERM U1030, Radiothérapie Moléculaire et Innovations Thérapeutiques, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
| | - Eloise Lopes
- INSERM U1030, Radiothérapie Moléculaire et Innovations Thérapeutiques, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
| | - Céline Clémenson
- INSERM U1030, Radiothérapie Moléculaire et Innovations Thérapeutiques, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
| | - Michele Mondini
- INSERM U1030, Radiothérapie Moléculaire et Innovations Thérapeutiques, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
| | - Lydia Meziani
- INSERM U1030, Radiothérapie Moléculaire et Innovations Thérapeutiques, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
| | - Yizhou Zhan
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, China
| | - Chengbing Zeng
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, China
| | - Qingxin Cai
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, China
| | - Daphne Morel
- Department of Radiation Oncology, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
- INSERM U1030, Radiothérapie Moléculaire et Innovations Thérapeutiques, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
| | - Roger Sun
- Faculté de Médecine, Université Paris-Saclay, 94270 Kremlin-Bicêtre, France
- Department of Radiation Oncology, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
- INSERM U1030, Radiothérapie Moléculaire et Innovations Thérapeutiques, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
| | - Pierre-Antoine Laurent
- Faculté de Médecine, Université Paris-Saclay, 94270 Kremlin-Bicêtre, France
- Department of Radiation Oncology, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
- INSERM U1030, Radiothérapie Moléculaire et Innovations Thérapeutiques, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
| | - Monica Mangoni
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences“Mario Serio” University of Florence, 50134 Florence, Italy
| | - Vanessa Di Cataldo
- Radiation Oncology Unit, Azienda Ospedaliero Universitaria Careggi, 50134 Florence, Italy
| | - Chiara Arilli
- Medical Physics Unit, Azienda Ospedaliero-Universitaria Careggi, 50134 Florence, Italy
| | - Maike Trommer
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
- Center for Integrated Oncology (CIO), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- Olivia Newton-John Cancer Wellness & Research Centre, Austin Health, Department of Radiation Oncology, HEIDELBERG VIC 3084, Melbourne, Australia
| | - Simone Wegen
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
- Center for Integrated Oncology (CIO), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Sebastian Neppl
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
| | - Rachel P. Riechelmann
- Department of Clinical Oncology, AC Camargo Cancer Center, São Paulo 01509-900, Brazil
| | - Marcos P. Camandaroba
- Department of Clinical Oncology, AC Camargo Cancer Center, São Paulo 01509-900, Brazil
| | - Elson Santos Neto
- Department of Radiation Oncology, AC Camargo Cancer Center, São Paulo 01509-001, Brazil
| | | | - Nicola Segata
- Department of Computational, Cellular and Integrative Biology, University of Trento, 38123 Trento, Italy
- IEO, Istituto Europeo di Oncologia IRCCS, 20139 Milan, Italy
| | - Peter Holicek
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
- Sotio Biotech,19000 Prague, Czech Republic
| | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA 19111-2497, USA
| | - Buqué Aitziber
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Carolina Alves Costa Silva
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR 1015, Equipe Labellisée—Ligue Nationale contre le Cancer, 94805 Villejuif, France
| | - Lisa Derosa
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif Cedex, France
- Faculté de Médecine, Université Paris-Saclay, 94270 Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR 1015, Equipe Labellisée—Ligue Nationale contre le Cancer, 94805 Villejuif, France
- CICBT1428, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, UMS AMMICa, Gustave Roussy Cancer Campus, 94805 Villejuif, France
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée – Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, 75006 Paris, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France
| | - Chuangzhen Chen
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, China
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif Cedex, France
- Faculté de Médecine, Université Paris-Saclay, 94270 Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR 1015, Equipe Labellisée—Ligue Nationale contre le Cancer, 94805 Villejuif, France
- CICBT1428, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
| | - Eric Deutsch
- Faculté de Médecine, Université Paris-Saclay, 94270 Kremlin-Bicêtre, France
- Department of Radiation Oncology, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
- INSERM U1030, Radiothérapie Moléculaire et Innovations Thérapeutiques, Gustave Roussy Cancer Campus (GRCC), 94805 Villejuif, France
| |
Collapse
|
8
|
Bravo-San Pedro JM, Aranda F, Buqué A, Galluzzi L. Animal models of disease: Achievements and challenges. Methods Cell Biol 2025; 192:xv-xxi. [PMID: 39863396 DOI: 10.1016/s0091-679x(25)00026-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
Affiliation(s)
- José Manuel Bravo-San Pedro
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | - Aitziber Buqué
- Fox Chase Cancer Center, Philadelphia, PA, United States.
| | | |
Collapse
|
9
|
Catanzaro E, Beltrán-Visiedo M, Galluzzi L, Krysko DV. Immunogenicity of cell death and cancer immunotherapy with immune checkpoint inhibitors. Cell Mol Immunol 2025; 22:24-39. [PMID: 39653769 PMCID: PMC11685666 DOI: 10.1038/s41423-024-01245-8] [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: 10/17/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024] Open
Abstract
While immunotherapy with immune checkpoint inhibitors (ICIs) has revolutionized the clinical management of various malignancies, a large fraction of patients are refractory to ICIs employed as standalone therapeutics, necessitating the development of combinatorial treatment strategies. Immunogenic cell death (ICD) inducers have attracted considerable interest as combinatorial partners for ICIs, at least in part owing to their ability to initiate a tumor-targeting adaptive immune response. However, compared with either approach alone, combinatorial regimens involving ICD inducers and ICIs have not always shown superior clinical activity. Here, we discuss accumulating evidence on the therapeutic interactions between ICD inducers and immunotherapy with ICIs in oncological settings, identify key factors that may explain discrepancies between preclinical and clinical findings, and propose strategies that address existing challenges to increase the efficacy of these combinations in patients with cancer.
Collapse
Affiliation(s)
- Elena Catanzaro
- Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Manuel Beltrán-Visiedo
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| | - Dmitri V Krysko
- Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.
| |
Collapse
|
10
|
Galassi C, Chan TA, Vitale I, Galluzzi L. The hallmarks of cancer immune evasion. Cancer Cell 2024; 42:1825-1863. [PMID: 39393356 DOI: 10.1016/j.ccell.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/27/2024] [Accepted: 09/16/2024] [Indexed: 10/13/2024]
Abstract
According to the widely accepted "three Es" model, the host immune system eliminates malignant cell precursors and contains microscopic neoplasms in a dynamic equilibrium, preventing cancer outgrowth until neoplastic cells acquire genetic or epigenetic alterations that enable immune escape. This immunoevasive phenotype originates from various mechanisms that can be classified under a novel "three Cs" conceptual framework: (1) camouflage, which hides cancer cells from immune recognition, (2) coercion, which directly or indirectly interferes with immune effector cells, and (3) cytoprotection, which shields malignant cells from immune cytotoxicity. Blocking the ability of neoplastic cells to evade the host immune system is crucial for increasing the efficacy of modern immunotherapy and conventional therapeutic strategies that ultimately activate anticancer immunosurveillance. Here, we review key hallmarks of cancer immune evasion under the "three Cs" framework and discuss promising strategies targeting such immunoevasive mechanisms.
Collapse
Affiliation(s)
- Claudia Galassi
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - 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
| | - Ilio Vitale
- Italian Institute for Genomic Medicine, c/o IRCSS Candiolo, Torino, Italy; Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medicine, 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; Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
11
|
García-Torralba E, Galluzzi L, Buqué A. Prognostic value of atypical B cells in breast cancer. Trends Cancer 2024; 10:990-991. [PMID: 39353814 DOI: 10.1016/j.trecan.2024.09.009] [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: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
The impact of tumor-infiltrating B cells on breast cancer (BRCA) outcomes remains poorly understood. Recent findings from Yang et al. identify an atypical, clonally expanded population of activated Fc receptor-like 4 (FCRL4)+ B cells that is associated with improved overall survival in patients affected by various tumor types, including BRCA.
Collapse
Affiliation(s)
- Esmeralda García-Torralba
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA; Department of Medical Oncology, Hospital Universitario Morales Meseguer, Murcia, Spain; Department of Medicine, Medical School, University of Murcia, Murcia, Spain; IMIB-Arrixaca, Murcia, Spain
| | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| | - Aitziber Buqué
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Carleton N, Lee S, Li R, Zou J, Brown DD, Hooda J, Chang A, Kumar R, Klei LR, Rigatti LH, Newsome J, John Mary DJS, Atkinson JM, West RE, Nolin TD, Oberly PJ, Huang Z, Poirier D, Diego EJ, Lucas PC, Tseng G, Lotze MT, McAuliffe PF, Zervantonakis IK, Oesterreich S, Lee AV. Systemic and local chronic inflammation and hormone disposition promote a tumor-permissive environment for breast cancer in older women. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.616978. [PMID: 39484485 PMCID: PMC11526964 DOI: 10.1101/2024.10.18.616978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Estrogen receptor positive (ER+) breast cancer is the most common subtype of breast cancer and is an age-related disease. The peak incidence of diagnosis occurs around age 70, even though these post-menopausal patients have low circulating levels of estradiol (E2). Despite the hormone sensitivity of age-related tumors, we have a limited understanding of the interplay between systemic and local hormones, chronic inflammation, and immune changes that contribute to the growth and development of these tumors. Here, we show that aged F344 rats treated with the dimethylbenz(a)anthracene / medroxyprogestrone acetate (DMBA/MPA) carcinogen develop more tumors at faster rates than their younger counterparts, suggesting that the aged environment promotes tumor initiation and impacts growth. Single-nuclei RNA-seq (snRNA-seq) of the tumors showed broad local immune dysfunction that was associated with circulating chronic inflammation. Across a broad cohort of specimens from patients with ER+ breast cancer and age-matched donors of normal breast tissue, we observe that even with an estrone (E1)-predominant estrogen disposition in the systemic circulation, tumors in older patients increase HSD17B7 expression to convert E1 to E2 in the tumor microenvironment (TME) and have local E2 levels similar to pre-menopausal patients. Concurrently, trackable increases in several chemokines, defined most notably by CCL2, promote a chronically inflamed but immune dysfunctional TME. This unique milieu in the aged TME, characterized by high local E2 and chemokine-enriched chronic inflammation, promotes both accumulation of tumor-associated macrophages (TAMs), which serve as signaling hubs, as well as polarization of TAMs towards a CD206+/PD-L1+, immunosuppressive phenotype. Pharmacologic targeting of estrogen signaling (either by HSD17B7 inhibition or with fulvestrant) and chemokine inflammation both decrease local E2 and prevent macrophage polarization. Overall, these findings suggest that chronic inflammation and hormonal disposition are critical contributors to the age-related nature of ER+ breast cancer development and growth and offer potential therapeutic insight to treat these patients. Translational Summary We uncover the unique underpinnings establishing how the systemic host environment contributes to the aged breast tumor microenvironment, characterized by high local estradiol and chronic inflammation with immune dysregulation, and show that targeting avenues of estrogen conversion and chronic inflammation work to restore anti-tumor immunity.
Collapse
|
13
|
Liu C, Fu L, Wang Y, Yang W. Influence of the gut microbiota on immune cell interactions and cancer treatment. J Transl Med 2024; 22:939. [PMID: 39407240 PMCID: PMC11476117 DOI: 10.1186/s12967-024-05709-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
The tumour microenvironment represents a novel frontier in oncological research. Over the past decade, accumulating evidence has underscored the importance of the tumour microenvironment (TME), including tumour cells, stromal cells, immune cells, and various secreted factors, which collectively influence tumour growth, invasion, and responses to therapeutic agents. Immune cells within the TME are now widely acknowledged to play pivotal roles in tumour development and treatment. While some perspectives have posited that immune cells within the TME facilitate tumour progression and confer resistance to therapeutic interventions, contrasting conclusions also exist. Affirmative and negative conclusions appear to be context dependent, and a unified consensus has yet to be reached. The burgeoning body of research on the relationship between the gut microbiota and tumours in recent years has led to a growing understanding. Most studies have indicated that specific components of the gut microbiota, such as unique bacterial communities or specific secretory factors, play diverse roles in regulating immune cells within the TME, thereby influencing the prognosis and outcomes of cancer treatments. A detailed understanding of these factors could provide novel insights into the TME and cancer therapy. In this study, we aimed to synthesise information on the interactions between the gut microbiota and immune cells within the TME, providing an in-depth exploration of the potential guiding implications for future cancer therapies.
Collapse
Affiliation(s)
- Chunxiao Liu
- Department of Gastroenterological Surgery, Hengqin Hospital, First Affiliated Hospital of Guangzhou Medical University, No. 118 Baoxing Road, Hengqin, Guangdong, 519031, China
| | - Lingfeng Fu
- Department of Gastroenterological Surgery, Hengqin Hospital, First Affiliated Hospital of Guangzhou Medical University, No. 118 Baoxing Road, Hengqin, Guangdong, 519031, China
| | - Yuxin Wang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, No. 1838, North Guangzhou Avenue, Guangzhou, Guangdong, 510515, China.
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Weijun Yang
- Department of Gastroenterological Surgery, Hengqin Hospital, First Affiliated Hospital of Guangzhou Medical University, No. 118 Baoxing Road, Hengqin, Guangdong, 519031, China.
| |
Collapse
|
14
|
Cordts SC, Yuki K, Henao Echeverri MF, Narasimhan B, Kuo CJ, Tang SKY. Microdissection tools to generate organoids for modeling the tumor immune microenvironment. MICROSYSTEMS & NANOENGINEERING 2024; 10:126. [PMID: 39251611 PMCID: PMC11385579 DOI: 10.1038/s41378-024-00756-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 09/11/2024]
Abstract
Patient-derived tumor organoids have emerged as promising models for predicting personalized drug responses in cancer therapy, but they typically lack immune components. Preserving the in vivo association between tumor cells and endogenous immune cells is critical for accurate testing of cancer immunotherapies. Mechanical dissection of tumor specimens into tumor fragments, as opposed to enzymatic digestion into single cells, is essential for maintaining these native tumor-immune cell spatial relationships. However, conventional mechanical dissection relying on manual mincing is time-consuming and irreproducible. This study describes two microdissection devices, the µDicer and µGrater, to facilitate the generation of intact tumor fragments from mouse B16 melanoma, a common model of human melanoma. The µDicer- and µGrater-cut tumor fragments were used to generate air‒liquid interface (ALI) organoids that copreserve tumor cells with infiltrating immune subsets without artificial reconstitution. The µDicer, consisting of a hexagonal array of silicon microblades, was employed to investigate the effect of organoid size. The viability of ALI organoid immune cells appeared insensitive to organoid sizes exceeding ~400 µm but diminished in organoids ~200 µm in size. The µGrater, consisting of an array of submillimeter holes in stainless steel, was employed to accelerate dissection. For the samples studied, the µGrater was 4.5 times faster than manual mincing. Compared with those generated by manual mincing, ALI organoids generated by the µGrater demonstrated similar viability, immune cell composition, and responses to anti-PD-1 immunotherapy. With further optimization, the µGrater holds potential for integration into clinical workflows to support the advancement of personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Seth C Cordts
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Kanako Yuki
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA
| | | | | | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA
| | - Sindy K Y Tang
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
15
|
Montégut L, Liu P, Zhao L, Pérez-Lanzón M, Chen H, Mao M, Zhang S, Derosa L, Naour JL, Lambertucci F, Mingoia S, Nogueira-Recalde U, Mena-Osuna R, Herranz-Montoya I, Djouder N, Baulande S, Pan H, Joseph A, Messaoudene M, Routy B, Fidelle M, Ben Ahmed T, Caron O, Busson P, Boulate D, Deschasaux-Tanguy M, Arnault N, Pol JG, Piaggio E, Touvier M, Zitvogel L, Delaloge S, Martins I, Kroemer G. Acyl-coenzyme a binding protein (ACBP) - a risk factor for cancer diagnosis and an inhibitor of immunosurveillance. Mol Cancer 2024; 23:187. [PMID: 39242519 PMCID: PMC11378439 DOI: 10.1186/s12943-024-02098-5] [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: 05/27/2024] [Accepted: 08/21/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND The plasma concentrations of acyl coenzyme A binding protein (ACBP, also known as diazepam-binding inhibitor, DBI, or 'endozepine') increase with age and obesity, two parameters that are also amongst the most important risk factors for cancer. METHODS We measured ACBP/DBI in the plasma from cancer-free individuals, high-risk patients like the carriers of TP53 or BRCA1/2 mutations, and non-syndromic healthy subjects who later developed cancer. In mice, the neutralization of ACBP/DBI was used in models of non-small cell lung cancer (NSCLC) and breast cancer development and as a combination treatment with chemoimmunotherapy (chemotherapy + PD-1 blockade) in the context of NSCLC and sarcomas. The anticancer T cell response upon ACBP/DBI neutralization was characterized by flow cytometry and single-cell RNA sequencing. RESULTS Circulating levels of ACBP/DBI were higher in patients with genetic cancer predisposition (BRCA1/2 or TP53 germline mutations) than in matched controls. In non-syndromic cases, high ACBP/DBI levels were predictive of future cancer development, and especially elevated in patients who later developed lung cancer. In preclinical models, ACBP/DBI neutralization slowed down breast cancer and NSCLC development and enhanced the efficacy of chemoimmunotherapy in NSCLC and sarcoma models. When combined with chemoimmunotherapy, the neutralizing monoclonal antibody against ACBP/DBI reduced the frequency of regulatory T cells in the tumor bed, modulated the immune checkpoint profile, and increased activation markers. CONCLUSION These findings suggest that ACBP/DBI acts as an endogenous immune suppressor. We conclude that elevation of ACBP/DBI constitutes a risk factor for the development of cancer and that ACBP/DBI is an actionable target for improving cancer immunosurveillance.
Collapse
Affiliation(s)
- Léa Montégut
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, Paris, France
| | - Peng Liu
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, Villejuif, France
| | - Liwei Zhao
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, Villejuif, France
| | - María Pérez-Lanzón
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, Villejuif, France
| | - Hui Chen
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, Paris, France
| | - Misha Mao
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, Villejuif, France
| | - Shuai Zhang
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, Villejuif, France
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lisa Derosa
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, Paris, France
- Equipe Labellisée Par la Ligue Contre le Cancer, Inserm U1015, Gustave Roussy, Villejuif, France
| | - Julie Le Naour
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, Paris, France
| | - Flavia Lambertucci
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, Villejuif, France
| | - Silvia Mingoia
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Paris, France
- Department of Pharmacological sciences, University of Piemonte Orientale, Novara, Italia
| | - Uxía Nogueira-Recalde
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, Villejuif, France
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Profesor Novoa Santos, A Coruña, Spain
| | - Rafael Mena-Osuna
- Department of Translational Research, Institute Curie Research Center, INSERM U932, PSL Research University, Paris, France
| | - Irene Herranz-Montoya
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, CNIO, Madrid, 28029, Spain
| | - Nabil Djouder
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, CNIO, Madrid, 28029, Spain
| | - Sylvain Baulande
- Institut Curie Genomics of Excellence (ICGex) Platform, Institut Curie Research Center, PSL Research University, Paris, France
| | - Hui Pan
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, Paris, France
| | - Adrien Joseph
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Paris, France
- Service de Réanimation Médicale, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Meriem Messaoudene
- Axe cancer, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
| | - Bertrand Routy
- Axe cancer, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Hemato-oncology Division, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Canada
| | - Marine Fidelle
- Equipe Labellisée Par la Ligue Contre le Cancer, Inserm U1015, Gustave Roussy, Villejuif, France
- Pharmacology Department, Gustave Roussy, Villejuif, France
| | - Tarek Ben Ahmed
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Department of cancer Medicine, « INTERCEPTION » Program for Cancer Prevention, Institut Gustave-Roussy, Villejuif, France
| | - Olivier Caron
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Department of cancer Medicine, « INTERCEPTION » Program for Cancer Prevention, Institut Gustave-Roussy, Villejuif, France
| | - Pierre Busson
- CNRS UMR 9018-METSY, Gustave Roussy and Université Paris-Saclay, Villejuif, France
| | - David Boulate
- Department of Thoracic Surgery, Lung Transplantation and Esophageal Diseases, Hôpital Nord, Marseille, France
- Faculté des sciences médicales et paramédicales, Aix-Marseille Université, Marseille, France
- COMPutational Oncology and pharmacology, Centre de Recherche en Cancérologie de Marseille (CRCM), INRIA-INSERM, Marseille, France
| | - Mélanie Deschasaux-Tanguy
- INRAE, CNAM, Nutritional Epidemiology Research Team (EREN), Université Sorbonne Paris Nord and Université Paris Cité, Centre of Research in Epidemiology and StatisticS (CRESS), Inserm, Bobigny, F-93017, France
- Nutrition, Physical Activity And Cancer Research Network (NACRe Network), Jouy-en-Josas, France
| | - Nathalie Arnault
- INRAE, CNAM, Nutritional Epidemiology Research Team (EREN), Université Sorbonne Paris Nord and Université Paris Cité, Centre of Research in Epidemiology and StatisticS (CRESS), Inserm, Bobigny, F-93017, France
- Nutrition, Physical Activity And Cancer Research Network (NACRe Network), Jouy-en-Josas, France
| | - Jonathan G Pol
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, Villejuif, France
| | - Eliane Piaggio
- Department of Translational Research, Institute Curie Research Center, INSERM U932, PSL Research University, Paris, France
| | - Mathilde Touvier
- INRAE, CNAM, Nutritional Epidemiology Research Team (EREN), Université Sorbonne Paris Nord and Université Paris Cité, Centre of Research in Epidemiology and StatisticS (CRESS), Inserm, Bobigny, F-93017, France
- Nutrition, Physical Activity And Cancer Research Network (NACRe Network), Jouy-en-Josas, France
| | - Laurence Zitvogel
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, Paris, France
- Equipe Labellisée Par la Ligue Contre le Cancer, Inserm U1015, Gustave Roussy, Villejuif, France
- Department of Biology, Center of Clinical Investigations in Biotherapies of Cancer (CICBT), BIOTHERIS, Villejuif, France
| | - Suzette Delaloge
- Equipe Labellisée Par la Ligue Contre le Cancer, Inserm U1015, Gustave Roussy, Villejuif, France
- Department of cancer Medicine, « INTERCEPTION » Program for Cancer Prevention, Institut Gustave-Roussy, Villejuif, France
| | - Isabelle Martins
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, Villejuif, France.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Inserm U1138, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Institut, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
16
|
Yu J, Yan Y, Li S, Xu Y, Parolia A, Rizvi S, Wang W, Zhai Y, Xiao R, Li X, Liao P, Zhou J, Okla K, Lin H, Lin X, Grove S, Wei S, Vatan L, Hu J, Szumilo J, Kotarski J, Freeman ZT, Skala S, Wicha M, Cho KR, Chinnaiyan AM, Schon S, Wen F, Kryczek I, Wang S, Chen L, Zou W. Progestogen-driven B7-H4 contributes to onco-fetal immune tolerance. Cell 2024; 187:4713-4732.e19. [PMID: 38968937 PMCID: PMC11344674 DOI: 10.1016/j.cell.2024.06.012] [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: 07/19/2023] [Revised: 02/09/2024] [Accepted: 06/09/2024] [Indexed: 07/07/2024]
Abstract
Immune tolerance mechanisms are shared in cancer and pregnancy. Through cross-analyzing single-cell RNA-sequencing data from multiple human cancer types and the maternal-fetal interface, we found B7-H4 (VTCN1) is an onco-fetal immune tolerance checkpoint. We showed that genetic deficiency of B7-H4 resulted in immune activation and fetal resorption in allogeneic pregnancy models. Analogously, B7-H4 contributed to MPA/DMBA-induced breast cancer progression, accompanied by CD8+ T cell exhaustion. Female hormone screening revealed that progesterone stimulated B7-H4 expression in placental and breast cancer cells. Mechanistically, progesterone receptor (PR) bound to a newly identified -58 kb enhancer, thereby mediating B7-H4 transcription via the PR-P300-BRD4 axis. PR antagonist or BRD4 degrader potentiated immunotherapy in a murine B7-H4+ breast cancer model. Thus, our work unravels a mechanistic and biological connection of a female sex hormone (progesterone) to onco-fetal immune tolerance via B7-H4 and suggests that the PR-P300-BRD4 axis is targetable for treating B7-H4+ cancer.
Collapse
Affiliation(s)
- Jiali Yu
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Yijian Yan
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Shasha Li
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Ying Xu
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Abhijit Parolia
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Syed Rizvi
- Department of Chemical Engineering, University of Michigan School of Engineering, Ann Arbor, MI, USA
| | - Weichao Wang
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Yiwen Zhai
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Rongxin Xiao
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Xiong Li
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Peng Liao
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Jiajia Zhou
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Karolina Okla
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA; Department of Oncological Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
| | - Heng Lin
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Xun Lin
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Sara Grove
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Shuang Wei
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Linda Vatan
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Jiantao Hu
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Justyna Szumilo
- Department of Clinical Pathomorphology, Medical University of Lublin, Lublin, Poland
| | - Jan Kotarski
- Department of Oncological Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
| | - Zachary T Freeman
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Stephanie Skala
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Max Wicha
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Kathleen R Cho
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Samantha Schon
- Department of Obstetrics and Gynecology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Fei Wen
- Department of Chemical Engineering, University of Michigan School of Engineering, Ann Arbor, MI, USA
| | - Ilona Kryczek
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Shaomeng Wang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lieping Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Weiping Zou
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA; Graduate Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
17
|
Ye J, Baer JM, Faget DV, Morikis VA, Ren Q, Melam A, Delgado AP, Luo X, Bagchi SM, Belle JI, Campos E, Friedman M, Veis DJ, Knudsen ES, Witkiewicz AK, Powers S, Longmore GD, DeNardo DG, Stewart SA. Senescent CAFs Mediate Immunosuppression and Drive Breast Cancer Progression. Cancer Discov 2024; 14:1302-1323. [PMID: 38683161 PMCID: PMC11216870 DOI: 10.1158/2159-8290.cd-23-0426] [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: 04/12/2023] [Revised: 01/26/2024] [Accepted: 03/08/2024] [Indexed: 05/01/2024]
Abstract
The tumor microenvironment (TME) profoundly influences tumorigenesis, with gene expression in the breast TME capable of predicting clinical outcomes. The TME is complex and includes distinct cancer-associated fibroblast (CAF) subtypes whose contribution to tumorigenesis remains unclear. Here, we identify a subset of myofibroblast CAFs (myCAF) that are senescent (senCAF) in mouse and human breast tumors. Utilizing the MMTV-PyMT;INK-ATTAC (INK) mouse model, we found that senCAF-secreted extracellular matrix specifically limits natural killer (NK) cell cytotoxicity to promote tumor growth. Genetic or pharmacologic senCAF elimination unleashes NK cell killing, restricting tumor growth. Finally, we show that senCAFs are present in HER2+, ER+, and triple-negative breast cancer and in ductal carcinoma in situ (DCIS) where they predict tumor recurrence. Together, these findings demonstrate that senCAFs are potently tumor promoting and raise the possibility that targeting them by senolytic therapy could restrain breast cancer development. Significance: senCAFs limit NK cell-mediated killing, thereby contributing to breast cancer progression. Thus, targeting senCAFs could be a clinically viable approach to limit tumor progression. See related article by Belle et al., p. 1324.
Collapse
Affiliation(s)
- Jiayu Ye
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John M. Baer
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Douglas V. Faget
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Vasilios A. Morikis
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Qihao Ren
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anupama Melam
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ana Paula Delgado
- Graduate Program in Genetics, Stony Brook University, Stony Brook, New York
| | - Xianmin Luo
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Satarupa Mullick Bagchi
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jad I. Belle
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Edward Campos
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael Friedman
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Deborah J. Veis
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Mo 63110, USA
| | | | | | - Scott Powers
- Department of Pathology and Cancer Center, Renaissance School of Medicine, Stony Brook, New York
| | - Gregory D. Longmore
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- ICCE Institute, Washington University School of Medicine, St Louis, MO
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David G. DeNardo
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- ICCE Institute, Washington University School of Medicine, St Louis, MO
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sheila A. Stewart
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- ICCE Institute, Washington University School of Medicine, St Louis, MO
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
18
|
Ceballos-Sanchez O, Navarro-López DE, Mejía-Méndez JL, Sanchez-Ante G, Rodríguez-González V, Sánchez-López AL, Sanchez-Martinez A, Duron-Torres SM, Juarez-Moreno K, Tiwari N, López-Mena ER. Enhancing antioxidant properties of CeO 2 nanoparticles with Nd 3+ doping: structural, biological, and machine learning insights. Biomater Sci 2024; 12:2108-2120. [PMID: 38450552 DOI: 10.1039/d3bm02107f] [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: 03/08/2024]
Abstract
The antioxidant capabilities of nanoparticles are contingent upon various factors, including their shape, size, and chemical composition. Herein, novel Nd-doped CeO2 nanoparticles were synthesized and the neodymium content was varied to investigate the synergistic impact on the antioxidant properties of CeO2 nanoparticles. Incorporating Nd3+ induced changes in lattice parameters and significantly altered the morphology from nanoparticles to nanorods. The biological activity of Nd-doped CeO2 was examined against pathogenic bacterial strains, breast cancer cell lines, and antioxidant models. The antibacterial and anticancer activities of nanoparticles were not observed, which could be associated with the Ce3+/Ce4+ ratio. Notably, the incorporation of neodymium improved the antioxidant capacity of CeO2. Machine learning techniques were employed to forecast the antioxidant activity to enhance understanding and predictive capabilities. Among these models, the random forest model exhibited the highest accuracy at 96.35%, establishing it as a robust computational tool for elucidating the biological behavior of Nd-doped CeO2 nanoparticles. This study presents the first exploration of the influence of Nd3+ on the structural, optical, and biological attributes of CeO2, contributing valuable insights and extending the application of machine learning in predicting the therapeutic efficacy of inorganic nanomaterials.
Collapse
Affiliation(s)
- Oscar Ceballos-Sanchez
- Universidad de Guadalajara, Centro Universitario de Ciencias Exactas e Ingenierias (CUCEI), Departamento de Ingenieria de Proyectos, Av. José Guadalupe Zuno # 48, Industrial Los Belenes, Zapopan, Jalisco, 45157, Mexico.
| | - Diego E Navarro-López
- Tecnologico de Monterrey, Escuela de ingeniería y Ciencias, Av. Gral. Ramón Corona No 2514, Colonia Nuevo México, Zapopan, Jalisco, 45121, Mexico
| | - Jorge L Mejía-Méndez
- Departamento de Ciencias Químico-Biológicas, Universidad de las Américas Puebla, Santa Catarina Mártir s/n, 72810 Cholula, Puebla, Mexico
| | - Gildardo Sanchez-Ante
- Tecnologico de Monterrey, Escuela de ingeniería y Ciencias, Av. Gral. Ramón Corona No 2514, Colonia Nuevo México, Zapopan, Jalisco, 45121, Mexico
| | - Vicente Rodríguez-González
- División de Materiales Avanzados, IPICYT, Instituto Potosino de Investigación Científica y Tecnológica, San Luis Potosí, S.L.P., Mexico
| | - Angélica Lizeth Sánchez-López
- Tecnologico de Monterrey, Escuela de ingeniería y Ciencias, Av. Gral. Ramón Corona No 2514, Colonia Nuevo México, Zapopan, Jalisco, 45121, Mexico
| | - Araceli Sanchez-Martinez
- Universidad de Guadalajara, Centro Universitario de Ciencias Exactas e Ingenierias (CUCEI), Departamento de Ingenieria de Proyectos, Av. José Guadalupe Zuno # 48, Industrial Los Belenes, Zapopan, Jalisco, 45157, Mexico.
| | - Sergio M Duron-Torres
- Unidad Académica de Ciencias Químicas, Universidad Autónoma de Zacatecas, Campus Siglo XXI, Carretera Zacatecas, Guadalajara Km 6, Ejido La Escondida, 98160, Zacatecas, Mexico
| | - Karla Juarez-Moreno
- Centro de Física Aplicada y Tecnología Avanzada (CFATA), Universidad Nacional Autónoma de México (UNAM), Querétaro, QRO 76230, Mexico
| | - Naveen Tiwari
- Centro Singular de Investigación en Química Biológica y Materiales Moleculares (CIQUS), C/Jenaro de la Fuente s/n, Campus Vida, Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Edgar R López-Mena
- Tecnologico de Monterrey, Escuela de ingeniería y Ciencias, Av. Gral. Ramón Corona No 2514, Colonia Nuevo México, Zapopan, Jalisco, 45121, Mexico
| |
Collapse
|
19
|
Perez-Lanzon M, Maiuri MC, Lopez-Otin C, Kroemer G. Preclinical models of breast cancer: B6BC, a transplantable hormone receptor-positive C57BL/6 mouse cell line. Genes Immun 2024; 25:172-174. [PMID: 38218998 DOI: 10.1038/s41435-023-00241-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/24/2023] [Accepted: 12/07/2023] [Indexed: 01/15/2024]
Affiliation(s)
- Maria Perez-Lanzon
- Centre de Recherche des Cordeliers, Équipe 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, UMS AMICCa, Gustave Roussy, Villejuif, France
| | - Maria Chiara Maiuri
- Centre de Recherche des Cordeliers, Équipe 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, UMS AMICCa, Gustave Roussy, Villejuif, France.
- Department of Molecular Medicine and Medical Biotechnologies, University of Napoli Federico II, Napoli, Italy.
| | - Carlos Lopez-Otin
- Departamento de Bioquimica y Biologia Molecular, Instituto Universitario de Oncologia (IUOPA), Universidad de Oviedo, Oviedo, Spain
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Équipe 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, UMS AMICCa, Gustave Roussy, Villejuif, France.
- Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, France-HP, Paris, France.
| |
Collapse
|
20
|
Bravo-San Pedro JM, Aranda F, Buqué A, Galluzzi L. Preface. Methods Cell Biol 2024; 185:xvii-xxiv. [PMID: 38556455 DOI: 10.1016/s0091-679x(24)00112-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Affiliation(s)
- José Manuel Bravo-San Pedro
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Fernando Aranda
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Aitziber Buqué
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States
| |
Collapse
|
21
|
Liu P, Zhao L, Zitvogel L, Kepp O, Kroemer G. Immunogenic cell death (ICD) enhancers-Drugs that enhance the perception of ICD by dendritic cells. Immunol Rev 2024; 321:7-19. [PMID: 37596984 DOI: 10.1111/imr.13269] [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: 07/17/2023] [Revised: 08/06/2023] [Accepted: 08/07/2023] [Indexed: 08/21/2023]
Abstract
The search for immunostimulatory drugs applicable to cancer immunotherapy may profit from target-agnostic methods in which agents are screened for their functional impact on immune cells cultured in vitro without any preconceived idea on their mode of action. We have built a synthetic mini-immune system in which stressed and dying cancer cells (derived from standardized cell lines) are confronted with dendritic cells (DCs, derived from immortalized precursors) and CD8+ T-cell hybridoma cells expressing a defined T-cell receptor. Using this system, we can identify three types of immunostimulatory drugs: (i) pharmacological agents that stimulate immunogenic cell death (ICD) of malignant cells; (ii) drugs that act on DCs to enhance their response to ICD; and (iii) drugs that act on T cells to increase their effector function. Here, we focus on strategies to develop drugs that enhance the perception of ICD by DCs and to which we refer as "ICD enhancers." We discuss examples of ICD enhancers, including ligands of pattern recognition receptors (exemplified by TLR3 ligands that correct the deficient function of DCs lacking FPR1) and immunometabolic modifiers (exemplified by hexokinase-2 inhibitors), as well as methods for target deconvolution applicable to the mechanistic characterization of ICD enhancers.
Collapse
Affiliation(s)
- Peng Liu
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Liwei Zhao
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Laurence Zitvogel
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Gustave Roussy, ClinicObiome, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Oliver Kepp
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| |
Collapse
|
22
|
Bravo-San Pedro JM, Aranda F, Buqué A, Galluzzi L. Animal models of disease: Achievements and challenges. Methods Cell Biol 2024; 188:xv-xxi. [PMID: 38880531 DOI: 10.1016/s0091-679x(24)00164-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Affiliation(s)
- José Manuel Bravo-San Pedro
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Fernando Aranda
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Aitziber Buqué
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States.
| |
Collapse
|
23
|
Pouget JP, Chan TA, Galluzzi L, Constanzo J. Radiopharmaceuticals as combinatorial partners for immune checkpoint inhibitors. Trends Cancer 2023; 9:968-981. [PMID: 37612188 PMCID: PMC11311210 DOI: 10.1016/j.trecan.2023.07.014] [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: 07/04/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/25/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of multiple cancer types. However, only a fraction of patients with cancer responds to ICIs employed as stand-alone therapeutics, calling for the development of safe and effective combinatorial regimens to extend the benefits of ICIs to a larger patient population. In addition to exhibiting a good safety and efficacy profile, targeted radionuclide therapy (TRT) with radiopharmaceuticals that specifically accumulate in the tumor microenvironment has been associated with promising immunostimulatory effects that (at least in preclinical cancer models) provide a robust platform for the development of TRT/ICI combinations. We discuss preclinical and clinical findings suggesting that TRT stands out as a promising partner for the development of safe and efficient combinatorial regimens involving ICIs.
Collapse
Affiliation(s)
- Jean-Pierre Pouget
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France.
| | - Timothy A Chan
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; National Center for Regenerative Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Centre, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| | - Julie Constanzo
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| |
Collapse
|
24
|
Zhao L, Liu P, Mao M, Zhang S, Bigenwald C, Dutertre CA, Lehmann CHK, Pan H, Paulhan N, Amon L, Buqué A, Yamazaki T, Galluzzi L, Kloeckner B, Silvin A, Pan Y, Chen H, Tian AL, Ly P, Dudziak D, Zitvogel L, Kepp O, Kroemer G. BCL2 Inhibition Reveals a Dendritic Cell-Specific Immune Checkpoint That Controls Tumor Immunosurveillance. Cancer Discov 2023; 13:2448-2469. [PMID: 37623817 PMCID: PMC7615270 DOI: 10.1158/2159-8290.cd-22-1338] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 07/13/2023] [Accepted: 08/23/2023] [Indexed: 08/26/2023]
Abstract
We developed a phenotypic screening platform for the functional exploration of dendritic cells (DC). Here, we report a genome-wide CRISPR screen that revealed BCL2 as an endogenous inhibitor of DC function. Knockout of BCL2 enhanced DC antigen presentation and activation as well as the capacity of DCs to control tumors and to synergize with PD-1 blockade. The pharmacologic BCL2 inhibitors venetoclax and navitoclax phenocopied these effects and caused a cDC1-dependent regression of orthotopic lung cancers and fibrosarcomas. Thus, solid tumors failed to respond to BCL2 inhibition in mice constitutively devoid of cDC1, and this was reversed by the infusion of DCs. Moreover, cDC1 depletion reduced the therapeutic efficacy of BCL2 inhibitors alone or in combination with PD-1 blockade and treatment with venetoclax caused cDC1 activation, both in mice and in patients. In conclusion, genetic and pharmacologic BCL2 inhibition unveils a DC-specific immune checkpoint that restrains tumor immunosurveillance. SIGNIFICANCE BCL2 inhibition improves the capacity of DCs to stimulate anticancer immunity and restrain cancer growth in an immunocompetent context but not in mice lacking cDC1 or mature T cells. This study indicates that BCL2 blockade can be used to sensitize solid cancers to PD-1/PD-L1-targeting immunotherapy. This article is featured in Selected Articles from This Issue, p. 2293.
Collapse
Affiliation(s)
- Liwei Zhao
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Peng Liu
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Misha Mao
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
- Surgical Oncology Department, Sir Run Run Shaw Hospital, Zhejiang University
| | - Shuai Zhang
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
- Department of Respiratory and Critical care Medicine, Union Hospital,Wuhan
| | - Camille Bigenwald
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Charles-Antoine Dutertre
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Christian H. K. Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Medical Immunology Campus Erlangen (MICE), Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
- Comprehensive Cancer Center Erlangen - European Metropolitan Area of Nuremberg, Erlangen, Germany
| | - Hui Pan
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
| | - Nicolas Paulhan
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
- Comprehensive Cancer Center Erlangen - European Metropolitan Area of Nuremberg, Erlangen, Germany
| | - Aitziber Buqué
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - 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
| | - Benoit Kloeckner
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Aymeric Silvin
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Yuhong Pan
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
| | - Hui Chen
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
| | - Ai-Ling Tian
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
| | - Pierre Ly
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Medical Immunology Campus Erlangen (MICE), Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
- Comprehensive Cancer Center Erlangen - European Metropolitan Area of Nuremberg, Erlangen, Germany
| | - Laurence Zitvogel
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Oliver Kepp
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, 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
|
25
|
Hahn AW, Venkatesh N, Msaouel P, McQuade JL. The Influence of Obesity on Outcomes with Immune Checkpoint Blockade: Clinical Evidence and Potential Biological Mechanisms. Cells 2023; 12:2551. [PMID: 37947629 PMCID: PMC10649394 DOI: 10.3390/cells12212551] [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: 09/15/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Immune checkpoint blockade (ICB) is a mainstay of treatment for advanced cancer, yet tumor response and host toxicity are heterogenous in those patients who receive ICB. There is growing interest in understanding how host factors interact with tumor intrinsic properties and the tumor microenvironment to influence the therapeutic index with ICB. Obesity, defined by body mass index, is a host factor associated with improved outcomes in select cancers when treated with ICB. While the biological mechanism for this obesity paradox is not fully understood, pre-clinical and translational studies suggest obesity may potentially impact tumor metabolism, inflammation, and angiogenesis. Herein, we summarize clinical studies that support an obesity paradox with ICB, explore potential biological mechanisms that may account for the obesity paradox, and address methodological challenges to consider when studying obesity and treatment outcomes.
Collapse
Affiliation(s)
- Andrew W. Hahn
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Neha Venkatesh
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Pavlos Msaouel
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer L. McQuade
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
26
|
Kepp O, Liu P, Kroemer G, Galluzzi L. A mitochondrial checkpoint to adaptive anticancer immunity. Oncoimmunology 2023; 12:2271693. [PMID: 37860277 PMCID: PMC10583618 DOI: 10.1080/2162402x.2023.2271693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/12/2023] [Indexed: 10/21/2023] Open
Abstract
BCL2 robustly preserves mitochondrial integrity, hence inhibiting innate immune signaling and apoptotic cell death in several cell types. Here, we comment on our recent data demonstrating that BCL2 also limits the ability of dendritic cells to elicit adaptive immune responses, lending support to a universal immunosuppressive function for the mitochondrial immune checkpoint.
Collapse
Affiliation(s)
- Oliver Kepp
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
| | - Peng Liu
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, 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, APHP, Paris, France
| | - 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
|
27
|
Pérez-Lanzón M, Maiuri MC, Lopez-Otin C, Kroemer G. Mammary carcinoma: toward a realistic mouse model of incurable cancers. Oncoimmunology 2023; 12:2240613. [PMID: 37546695 PMCID: PMC10402843 DOI: 10.1080/2162402x.2023.2240613] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 08/08/2023] Open
Abstract
As long as breast cancer (BC) stays under immunosurveillance, it can be controlled by treatments eliciting anticancer immune responses. However, once BC escapes immunosurveillance, it becomes therapeutically uncontrollable. A paper in the Journal for ImmunoTherapy of Cancer describes a new hormone receptor-positive BC cell line generating incurable tumors in C57BL/6 mice.
Collapse
Affiliation(s)
- Maria Pérez-Lanzón
- Centre de Recherche des Cordeliers, Équipe 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, UMS AMICCa, Gustave Roussy, Villejuif, France
| | - Maria Chiara Maiuri
- Centre de Recherche des Cordeliers, Équipe 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, UMS AMICCa, Gustave Roussy, Villejuif, France
- Department of Molecular Medicine and Medical Biotechnologies, University of Napoli Federico II, Napoli, Italy
| | - Carlos Lopez-Otin
- Departamento de Bioquimica Y Biologia Molecular, Instituto Universitario de Oncologia (IUOPA), Universidad de Oviedo, Oviedo, Spain
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Équipe 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, UMS AMICCa, Gustave Roussy, Villejuif, France
- Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, France-HP, Paris, France
| |
Collapse
|
28
|
Ollivier L, Moreau Bachelard C, Renaud E, Dhamelincourt E, Lucia F. The abscopal effect of immune-radiation therapy in recurrent and metastatic cervical cancer: a narrative review. Front Immunol 2023; 14:1201675. [PMID: 37539054 PMCID: PMC10394237 DOI: 10.3389/fimmu.2023.1201675] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/30/2023] [Indexed: 08/05/2023] Open
Abstract
Despite human papillomavirus vaccination and screening, in about 5% of cases, cervical cancer (CC) is discovered at an initial metastatic stage. Moreover, nearly one-third of patients with locally advanced CC (LACC) will have a recurrence of their disease during follow-up. At the stage of recurrent or metastatic CC, there are very few treatment options. They are considered incurable with a very poor prognosis. For many years, the standard of care was the combination of platinum-based drug and paclitaxel with the possible addition of bevacizumab. The most recent years have seen the development of the use of immune checkpoint inhibitors (ICIs) (pembrolizumab, cemiplimab and others) in patients with CC. They have shown long term responses with improved overall survival of patients in 1st line (in addition to chemotherapy) or 2nd line (as monotherapy) treatment. Another emerging drug is tisotumab vedotin, an antibody-drug conjugate targeting tissue factor. Radiation therapy (RT) often has a limited palliative indication in metastatic cancers. However, it has been observed that RT can induce tumor shrinkage both in distant metastatic tumors beyond the radiation field and in primary irradiated tumors. This is a rarely observed phenomenon, called abscopal effect, which is thought to be related to the immune system and allows a tumor response throughout the body. It would be the activation of the immune system induced by the irradiation of cancer cells that would lead to a specific type of apoptosis, the immunogenic cell death. Today, there is a growing consensus that combining RT with ICIs may boost abscopal response or cure rates for various cancers. Here we will review the potential abscopal effect of immune-radiation therapy in metastatic cervical cancer.
Collapse
Affiliation(s)
- Luc Ollivier
- Department of Radiation Oncology, Institut De Cancérologie De L’Ouest (ICO), Saint-Herblain, France
| | | | - Emmanuelle Renaud
- Department of Medical Oncology, CHRU Morvan, University Hospital, Brest, France
| | | | - Francois Lucia
- Radiation Oncology Department, University Hospital, Brest, France
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| |
Collapse
|
29
|
Klapp V, Álvarez-Abril B, Leuzzi G, Kroemer G, Ciccia A, Galluzzi L. The DNA Damage Response and Inflammation in Cancer. Cancer Discov 2023; 13:1521-1545. [PMID: 37026695 DOI: 10.1158/2159-8290.cd-22-1220] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/27/2023] [Accepted: 02/23/2023] [Indexed: 04/08/2023]
Abstract
Genomic stability in normal cells is crucial to avoid oncogenesis. Accordingly, multiple components of the DNA damage response (DDR) operate as bona fide tumor suppressor proteins by preserving genomic stability, eliciting the demise of cells with unrepairable DNA lesions, and engaging cell-extrinsic oncosuppression via immunosurveillance. That said, DDR sig-naling can also favor tumor progression and resistance to therapy. Indeed, DDR signaling in cancer cells has been consistently linked to the inhibition of tumor-targeting immune responses. Here, we discuss the complex interactions between the DDR and inflammation in the context of oncogenesis, tumor progression, and response to therapy. SIGNIFICANCE Accumulating preclinical and clinical evidence indicates that DDR is intimately connected to the emission of immunomodulatory signals by normal and malignant cells, as part of a cell-extrinsic program to preserve organismal homeostasis. DDR-driven inflammation, however, can have diametrically opposed effects on tumor-targeting immunity. Understanding the links between the DDR and inflammation in normal and malignant cells may unlock novel immunotherapeutic paradigms to treat cancer.
Collapse
Affiliation(s)
- Vanessa Klapp
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Tumor Stroma Interactions, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Beatriz Álvarez-Abril
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Department of Hematology and Oncology, Hospital Universitario Morales Meseguer, Murcia, Spain
| | - Giuseppe Leuzzi
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, New York
- Herbert Irving Comprehensive Cancer Center, New York, New York
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, New York
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le Cancer, Université de Paris, 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
| | - Alberto Ciccia
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, New York
- Herbert Irving Comprehensive Cancer Center, New York, New York
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, New York
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Sandra and Edward Meyer Cancer Center, New York, New York
- Caryl and Israel Englander Institute for Precision Medicine, New York, New York
| |
Collapse
|
30
|
Zimmermann A, Hofer SJ, Madeo F. Molecular targets of spermidine: implications for cancer suppression. Cell Stress 2023; 7:50-58. [PMID: 37431488 PMCID: PMC10320397 DOI: 10.15698/cst2023.07.281] [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: 05/15/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/12/2023] Open
Abstract
Spermidine is a ubiquitous, natural polyamine with geroprotective features. Supplementation of spermidine extends the lifespan of yeast, worms, flies, and mice, and dietary spermidine intake correlates with reduced human mortality. However, the crucial role of polyamines in cell proliferation has also implicated polyamine metabolism in neoplastic diseases, such as cancer. While depleting intracellular polyamine biosynthesis halts tumor growth in mouse models, lifelong external spermidine administration in mice does not increase cancer incidence. In contrast, a series of recent findings points to anti-neoplastic properties of spermidine administration in the context of immunotherapy. Various molecular mechanisms for the anti-aging and anti-cancer properties have been proposed, including the promotion of autophagy, enhanced translational control, and augmented mitochondrial function. For instance, spermidine allosterically activates mitochondrial trifunctional protein (MTP), a bipartite protein complex that mediates three of the four steps of mitochondrial fatty acid (β-oxidation. Through this action, spermidine supplementation is able to restore MTP-mediated mitochondrial respiratory capacity in naïve CD8+ T cells to juvenile levels and thereby improves T cell activation in aged mice. Here, we put this finding into the context of the previously described molecular target space of spermidine.
Collapse
Affiliation(s)
- Andreas Zimmermann
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Sebastian J. Hofer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| |
Collapse
|
31
|
Galluzzi L, Aryankalayil MJ, Coleman CN, Formenti SC. Emerging evidence for adapting radiotherapy to immunotherapy. Nat Rev Clin Oncol 2023:10.1038/s41571-023-00782-x. [PMID: 37280366 DOI: 10.1038/s41571-023-00782-x] [Citation(s) in RCA: 147] [Impact Index Per Article: 73.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2023] [Indexed: 06/08/2023]
Abstract
Immunotherapy has revolutionized the clinical management of many malignancies but is infrequently associated with durable objective responses when used as a standalone treatment approach, calling for the development of combinatorial regimens with superior efficacy and acceptable toxicity. Radiotherapy, the most commonly used oncological treatment, has attracted considerable attention as a combination partner for immunotherapy owing to its well-known and predictable safety profile, widespread clinical availability, and potential for immunostimulatory effects. However, numerous randomized clinical trials investigating radiotherapy-immunotherapy combinations have failed to demonstrate a therapeutic benefit compared with either modality alone. Such a lack of interaction might reflect suboptimal study design, choice of end points and/or administration of radiotherapy according to standard schedules and target volumes. Indeed, radiotherapy has empirically evolved towards radiation doses and fields that enable maximal cancer cell killing with manageable toxicity to healthy tissues, without much consideration of potential radiation-induced immunostimulatory effects. Herein, we propose the concept that successful radiotherapy-immunotherapy combinations might require modifications of standard radiotherapy regimens and target volumes to optimally sustain immune fitness and enhance the antitumour immune response in support of meaningful clinical benefits.
Collapse
Affiliation(s)
- 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
| | - Molykutty J Aryankalayil
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - C Norman Coleman
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
32
|
Terrisse S, Zitvogel L, Kroemer G. Impact of microbiota on breast cancer hormone therapy. Cell Stress 2023; 7:12-19. [PMID: 36926118 PMCID: PMC10012050 DOI: 10.15698/cst2023.03.277] [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: 02/14/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/12/2023] Open
Abstract
Recent observations indicate that the pathogenesis and prognosis of hormone-receptor breast cancer is not only dictated by the properties of the malignant cells but also by immune and microbial parameters. Thus, the immunosurveillance system retards the development of hormone-positive breast cancer and contributes to the therapeutic efficacy of estrogen receptor antagonists and aromatase inhibitors. Moreover, the anticancer immune response is profoundly modulated by the local and intestinal microbiota, which influences cancer cell-intrinsic signaling pathways, affects the composition and function of the immune infiltrate present in the tumor microenvironment and modulates the metabolism of estrogens. Indeed, specific bacteria in the gut produce enzymes that affect the enterohepatic cycle of estrogen metabolites, convert estrogens into androgens or generate estrogen-like molecules. The knowledge of these circuitries is in its infancy, calling for further in-depth analyses.
Collapse
Affiliation(s)
| | - Laurence Zitvogel
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France.,University Paris Saclay, Gif-sur-Yvette, France.,Gustave Roussy, ClinicObiome, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - 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
|
33
|
Abstract
Numerous mitochondrial constituents and metabolic products can function as damage-associated molecular patterns (DAMPs) and promote inflammation when released into the cytosol or extracellular milieu. Several safeguards are normally in place to prevent mitochondria from eliciting detrimental inflammatory reactions, including the autophagic disposal of permeabilized mitochondria. However, when the homeostatic capacity of such systems is exceeded or when such systems are defective, inflammatory reactions elicited by mitochondria can become pathogenic and contribute to the aetiology of human disorders linked to autoreactivity. In addition, inefficient inflammatory pathways induced by mitochondrial DAMPs can be pathogenic as they enable the establishment or progression of infectious and neoplastic disorders. Here we discuss the molecular mechanisms through which mitochondria control inflammatory responses, the cellular pathways that are in place to control mitochondria-driven inflammation and the pathological consequences of dysregulated inflammatory reactions elicited by mitochondrial DAMPs.
Collapse
Affiliation(s)
- Saverio Marchi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Emma Guilbaud
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Stephen W G Tait
- Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
| | - 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
|
34
|
Klapp V, Buqué A, Bloy N, Sato A, Yamazaki T, Zhou XK, Formenti SC, Galluzzi L, Petroni G. Cellular senescence in the response of HR + breast cancer to radiotherapy and CDK4/6 inhibitors. J Transl Med 2023; 21:110. [PMID: 36765430 PMCID: PMC9921325 DOI: 10.1186/s12967-023-03964-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 02/02/2023] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND Preclinical evidence from us and others demonstrates that the anticancer effects of cyclin-dependent kinase 4/6 (CDK4/6) inhibitors can be enhanced with focal radiation therapy (RT), but only when RT is delivered prior to (rather than after) CDK4/6 inhibition. Depending on tumor model, cellular senescence (an irreversible proliferative arrest that is associated with the secretion of numerous bioactive factors) has been attributed beneficial or detrimental effects on response to treatment. As both RT and CDK4/6 inhibitors elicit cellular senescence, we hypothesized that a differential accumulation of senescent cells in the tumor microenvironment could explain such an observation, i.e., the inferiority of CDK4/6 inhibition with palbociclib (P) followed by RT (P→RT) as compared to RT followed by palbociclib (RT→P). METHODS The impact of cellular senescence on the interaction between RT and P was assessed by harnessing female INK-ATTAC mice, which express a dimerizable form of caspase 8 (CASP8) under the promoter of cyclin dependent kinase inhibitor 2A (Cdkn2a, coding for p16Ink4), as host for endogenous mammary tumors induced by the subcutaneous implantation of medroxyprogesterone acetate (MPA, M) pellets combined with the subsequent oral administration of 7,12-dimethylbenz[a]anthracene (DMBA, D). This endogenous mouse model of HR+ mammary carcinogenesis recapitulates key immunobiological aspects of human HR+ breast cancer. Mice bearing M/D-driven tumors were allocated to RT, P or their combination in the optional presence of the CASP8 dimerizer AP20187, and monitored for tumor growth, progression-free survival and overall survival. In parallel, induction of senescence in vitro, in cultured human mammary hormone receptor (HR)+ adenocarcinoma MCF7 cells, triple negative breast carcinoma MDA-MB-231 cells and mouse HR+ mammary carcinoma TS/A cells treated with RT, P or their combination, was determined by colorimetric assessment of senescence-associated β-galactosidase activity after 3 or 7 days of treatment. RESULTS In vivo depletion of p16Ink4-expressing (senescent) cells ameliorated the efficacy of P→RT (but not that of RT→P) in the M/D-driven model of HR+ mammary carcinogenesis. Accordingly, P→RT induced higher levels of cellular senescence than R→TP in cultured human and mouse breast cancer cell lines. CONCLUSIONS Pending validation in other experimental systems, these findings suggest that a program of cellular senescence in malignant cells may explain (at least partially) the inferiority of P→RT versus RT→P in preclinical models of HR+ breast cancer.
Collapse
Affiliation(s)
- Vanessa Klapp
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Aitziber Buqué
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Norma Bloy
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Ai Sato
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Xi Kathy Zhou
- Healthcare Policy and Research, Weill Cornell Medical College, New York, NY, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - 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.
| | - Giulia Petroni
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| |
Collapse
|
35
|
Quantitative assessment of mitophagy in irradiated cancer cells. Methods Cell Biol 2023; 174:93-111. [PMID: 36710054 DOI: 10.1016/bs.mcb.2022.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Mitophagy is a finely regulated mechanism through which eukaryotic cells selectively dispose of supernumerary, permeabilized or otherwise damaged mitochondria through lysosomal degradation. Dysfunctional mitochondria are prone to release potentially cytotoxic factors including reactive oxygen species (ROS) and caspase activators, such as cytochrome c, somatic (CYCS). Thus, proficient mitophagic responses mediate prominent cytoprotective functions. Moreover, the rapid degradation of permeabilized mitochondria limits the release of mitochondrial components that may drive inflammatory reactions, such as mitochondrial DNA (mtDNA) and transcription factor A, mitochondrial (TFAM), implying that mitophagy also mediates potent anti-inflammatory effects. Here, we detail a simple, flow cytometry-assisted protocol for the specific measurement of mitophagic responses as driven by radiation therapy (RT) in mouse hormone receptor (HR)+ mammary carcinoma TS/A cells. With some variations, this method - which relies on the mitochondria-restricted expression of a fluorescent reporter that is sensitive to pH and hence changes excitation wavelength within lysosomes (mt-mKeima) - can be adapted to a variety of human and mouse cancer cell lines and/or straightforwardly implemented on fluorescence microscopy platforms.
Collapse
|
36
|
Sato A, Kraynak J, Marciscano AE, Galluzzi L. Radiation therapy: An old dog learning new tricks. Methods Cell Biol 2023; 174:xv-xxv. [PMID: 37039770 DOI: 10.1016/s0091-679x(23)00036-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
37
|
Carbonnier V, Le Naour J, Bachelot T, Vacchelli E, André F, Delaloge S, Kroemer G. Rs867228 in FPR1 accelerates the manifestation of luminal B breast cancer. Oncoimmunology 2023; 12:2189823. [PMID: 36970071 PMCID: PMC10038022 DOI: 10.1080/2162402x.2023.2189823] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
Abstract
Formyl peptide receptor-1 (FPR1) is a pathogen recognition receptor involved in the detection of bacteria, in the control of inflammation, as well as in cancer immunosurveillance. A single nucleotide polymorphism in FPR1, rs867228, provokes a loss-of-function phenotype. In a bioinformatic study performed on The Cancer Genome Atlas (TCGA), we observed that homo-or heterozygosity for rs867228 in FPR1 (which affects approximately one-third of the population across continents) accelerates age at diagnosis of specific carcinomas including luminal B breast cancer by 4.9 years. To validate this finding, we genotyped 215 patients with metastatic luminal B mammary carcinomas from the SNPs To Risk of Metastasis (SToRM) cohort. The first diagnosis of luminal B breast cancer occurred at an age of 49.2 years for individuals bearing the dysfunctional TT or TG alleles (n = 73) and 55.5 years for patients the functional GG alleles (n = 141), meaning that rs867228 accelerated the age of diagnosis by 6.3 years (p=0.0077, Mann & Whitney). These results confirm our original observation in an independent validation cohort. We speculate that it may be useful to include the detection of rs867228 in breast cancer screening campaigns for selectively increasing the frequency and stringency of examinations starting at a relatively young age.
Collapse
Affiliation(s)
- Vincent Carbonnier
- Equipe labellisée par la Ligue contrele cancer, Université de Paris, Sorbonne Université, Paris, France
| | - Julie Le Naour
- Equipe labellisée par la Ligue contrele cancer, Université de Paris, Sorbonne Université, Paris, France
| | - Thomas Bachelot
- Centre Léon Bérard, Département de Cancérologie Médicale, Lyon, France
| | - Erika Vacchelli
- Equipe labellisée par la Ligue contrele cancer, Université de Paris, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Fabrice André
- Université Paris Saclay, Faculty of Medicine Kremlin Bicêtre, Le Kremlin Bicêtre, France
- Department of Medical Oncology, INSERM U981, Gustave Roussy Cancer Campus, Villejuif, France
| | - Suzette Delaloge
- Department of Cancer Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| | - Guido Kroemer
- Equipe labellisée par la Ligue contrele cancer, Université de Paris, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- CONTACT Guido Kroemer Equipe labellisée par la Ligue contrele cancer, Université de Paris, Sorbonne Université, Centre de Recherche des Cordeliers, 15 rue de l’Ecole de Médecine, Paris75006, France
| |
Collapse
|
38
|
Mirjolet C, Baude J, Galluzzi L. Dual impact of radiation therapy on tumor-targeting immune responses. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 378:xiii-xxiv. [PMID: 37438022 DOI: 10.1016/s1937-6448(23)00114-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Affiliation(s)
- Céline Mirjolet
- Radiation Oncology Department, Preclinical Radiation Therapy and Radiobiology Unit, GF Leclerc Centre, Unicancer, Dijon, France; TIReCS Team, UMR INSERM 1231, Dijon, France.
| | - Jérémy Baude
- Radiation Oncology Department, Preclinical Radiation Therapy and Radiobiology Unit, GF Leclerc Centre, Unicancer, Dijon, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States.
| |
Collapse
|
39
|
Sato A, Kraynak J, Marciscano AE, Galluzzi L. Radiation therapy: An old dog learning new tricks. Methods Cell Biol 2023; 180:xv-xxv. [PMID: 37890936 DOI: 10.1016/s0091-679x(23)00166-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Affiliation(s)
- Ai Sato
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Jeffrey Kraynak
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Ariel E Marciscano
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States.
| |
Collapse
|
40
|
Abstract
Historically, cancer research and therapy have focused on malignant cells and their tumor microenvironment. However, the vascular, lymphatic and nervous systems establish long-range communication between the tumor and the host. This communication is mediated by metabolites generated by the host or the gut microbiota, as well by systemic neuroendocrine, pro-inflammatory and immune circuitries-all of which dictate the trajectory of malignant disease through molecularly defined biological mechanisms. Moreover, aging, co-morbidities and co-medications have a major impact on the development, progression and therapeutic response of patients with cancer. In this Perspective, we advocate for a whole-body 'ecological' exploration of malignant disease. We surmise that accumulating knowledge on the intricate relationship between the host and the tumor will shape rational strategies for systemic, bodywide interventions that will eventually improve tumor control, as well as quality of life, in patients with cancer.
Collapse
|
41
|
Tong L, Jiménez-Cortegana C, Tay AHM, Wickström S, Galluzzi L, Lundqvist A. NK cells and solid tumors: therapeutic potential and persisting obstacles. Mol Cancer 2022; 21:206. [PMID: 36319998 PMCID: PMC9623927 DOI: 10.1186/s12943-022-01672-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/10/2022] [Accepted: 09/24/2022] [Indexed: 11/05/2022] Open
Abstract
Natural killer (NK) cells, which are innate lymphocytes endowed with potent cytotoxic activity, have recently attracted attention as potential anticancer therapeutics. While NK cells mediate encouraging responses in patients with leukemia, the therapeutic effects of NK cell infusion in patients with solid tumors are limited. Preclinical and clinical data suggest that the efficacy of NK cell infusion against solid malignancies is hampered by several factors including inadequate tumor infiltration and persistence/activation in the tumor microenvironment (TME). A number of metabolic features of the TME including hypoxia as well as elevated levels of adenosine, reactive oxygen species, and prostaglandins negatively affect NK cell activity. Moreover, cancer-associated fibroblasts, tumor-associated macrophages, myeloid-derived suppressor cells, and regulatory T cells actively suppress NK cell-dependent anticancer immunity. Here, we review the metabolic and cellular barriers that inhibit NK cells in solid neoplasms as we discuss potential strategies to circumvent such obstacles towards superior therapeutic activity.
Collapse
Affiliation(s)
- Le Tong
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Carlos Jiménez-Cortegana
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Department of Medical Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Seville, Seville, Spain
| | - Apple H M Tay
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
- Department of Biological Science, Nanyang Technological University, Singapore, Singapore
| | - Stina Wickström
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - 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.
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
42
|
Montégut L, de Cabo R, Zitvogel L, Kroemer G. Science-Driven Nutritional Interventions for the Prevention and Treatment of Cancer. Cancer Discov 2022; 12:2258-2279. [PMID: 35997502 PMCID: PMC10749912 DOI: 10.1158/2159-8290.cd-22-0504] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/18/2022] [Accepted: 07/15/2022] [Indexed: 11/16/2022]
Abstract
In population studies, dietary patterns clearly influence the development, progression, and therapeutic response of cancers. Nonetheless, interventional dietary trials have had relatively little impact on the prevention and treatment of malignant disease. Standardization of nutritional interventions combined with high-level mode-of-action studies holds the promise of identifying specific entities and pathways endowed with antineoplastic properties. Here, we critically review the effects of caloric restriction and more specific interventions on macro- and micronutrients in preclinical models as well as in clinical studies. We place special emphasis on the prospect of using defined nutrition-relevant molecules to enhance the efficacy of established anticancer treatments. SIGNIFICANCE The avoidance of intrinsically hypercaloric and toxic diets contributes to the prevention and cure of cancer. In addition, specific diet-induced molecules such as ketone bodies and micronutrients, including specific vitamins, have drug-like effects that are clearly demonstrable in preclinical models, mostly in the context of immunotherapies. Multiple trials are underway to determine the clinical utility of such molecules.
Collapse
Affiliation(s)
- Léa Montégut
- Equipe labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, Université de Paris Cité, Sorbonne Université, Institut Universitaire de France, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Faculty of Medicine, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland
| | - Laurence Zitvogel
- Faculty of Medicine, Université Paris Saclay, Le Kremlin-Bicêtre, France
- Gustave Roussy Comprehensive Cancer Institute, ClinicObiome, Villejuif, France
- INSERM U1015, Paris, France
- Equipe labellisée par la Ligue contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) BIOTHERIS, Villejuif, France
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, Université de Paris Cité, Sorbonne Université, Institut Universitaire de France, Inserm U1138, 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
|
43
|
Hofer SJ, Kroemer G, Kepp O. Autophagy-inducing nutritional interventions in experimental and clinical oncology. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 373:125-158. [PMID: 36283765 DOI: 10.1016/bs.ircmb.2022.08.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Numerous pro-autophagic dietary interventions are being investigated for their potential cancer-preventive or therapeutic effects. This applies to different fasting regimens, methionine restriction and ketogenic diets. In addition, the supplementation of specific micronutrients such as nicotinamide (vitamin B3) or spermidine induces autophagy. In humans, leanness, plant-based diets (that may lead to partial methionine restriction) and high dietary uptake of spermidine are associated with a low incidence of cancers. Moreover, clinical trials have demonstrated the capacity of nicotinamide to prevent non-melanoma skin carcinogenesis. Multiple interventional trials are evaluating the capacity of autophagy-inducing regimens to improve the outcome of chemotherapy and immunotherapy. Here, we discuss the mechanistic underpinnings of autophagy induction by nutritional interventions, as well as the mechanisms through which autophagy induction in malignant or immune cells improves anticancer immunosurveillance.
Collapse
Affiliation(s)
- Sebastian J Hofer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Paris, France; Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Paris, France; Institut du Cancer Paris Carpem, Department of Biology, APHP, Hôpital Européen Georges Pompidou, Paris, France.
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Paris, France.
| |
Collapse
|
44
|
Hu Y, Bloy N, Elemento O, Buqué A. Nicotinamide drives T cell activation in the mammary tumor microenvironment. J Transl Med 2022; 20:251. [PMID: 35659314 PMCID: PMC9164530 DOI: 10.1186/s12967-022-03454-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 11/10/2022] Open
Abstract
Nicotinamide (NAM, a variant of vitamin B3) has recently been shown to accelerate the activation of human CD4+ and CD8+ T cells exposed to repeated CD3/CD28 agonism in vitro. Here, we demonstrate that T cells infiltrating mouse mammary carcinomas that are therapeutically controlled by NAM also express multiple markers of late-stage activation. Taken together, these findings lend additional support to the notion that the antineoplastic effects of NAM involve at least some degree of restored cancer immunosurveillance.
Collapse
Affiliation(s)
- Yang Hu
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
| | - Norma Bloy
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA.,Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Aitziber Buqué
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
45
|
Petrazzuolo A, Maiuri MC, Zitvogel L, Kroemer G, Kepp O. Trial Watch: combination of tyrosine kinase inhibitors (TKIs) and immunotherapy. Oncoimmunology 2022; 11:2077898. [PMID: 35655707 PMCID: PMC9154809 DOI: 10.1080/2162402x.2022.2077898] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The past decades witnessed the clinical employment of targeted therapies including but not limited to tyrosine kinase inhibitors (TKIs) that restrain a broad variety of pro-tumorigenic signals. TKIs can be categorized into (i) agents that directly target cancer cells, (ii) normalize angiogenesis or (iii) affect cells of the hematologic lineage. However, a clear distinction of TKIs based on this definition is limited by the fact that many TKIs designed to inhibit cancer cells have also effects on immune cells that are being discovered. Additionally, TKIs originally designed to target hematological cancers exhibit bioactivities on healthy cells of the same hematological lineage. TKIs have been described to improve immune recognition and cancer immunosurveillance, providing the scientific basis to combine TKIs with immunotherapy. Indeed, combination of TKIs with immunotherapy showed synergistic effects in preclinical models and clinical trials and some combinations of TKIs normalizing angiogenesis with immune checkpoint blocking antibodies have already been approved by the FDA for cancer therapy. However, the identification of appropriate drug combinations as well as optimal dosing and scheduling needs to be improved in order to obtain tangible progress in cancer care. This Trial Watch summarizes active clinical trials combining TKIs with various immunotherapeutic strategies to treat cancer patients.
Collapse
Affiliation(s)
- Adriana Petrazzuolo
- Team “Metabolism, Cancer & Immunity”, Centre de Recherche des Cordeliers, INSERM UMRS1138, Université Paris Cité, Sorbonne Université, Paris, France
- Cell Biology and Metabolomics platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - M. Chiara Maiuri
- Team “Metabolism, Cancer & Immunity”, Centre de Recherche des Cordeliers, INSERM UMRS1138, Université Paris Cité, Sorbonne Université, Paris, France
- Cell Biology and Metabolomics platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Laurence Zitvogel
- Faculty of Medicine, University Paris Saclay, Kremlin Bicêtre, France
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Institut National de la Santé et de la Recherche Medicale (INSERM) U1015, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) Biotheris 1428, Villejuif, France
| | - Guido Kroemer
- Team “Metabolism, Cancer & Immunity”, Centre de Recherche des Cordeliers, INSERM UMRS1138, Université Paris Cité, Sorbonne Université, Paris, France
- Cell Biology and Metabolomics platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Oliver Kepp
- Team “Metabolism, Cancer & Immunity”, Centre de Recherche des Cordeliers, INSERM UMRS1138, Université Paris Cité, Sorbonne Université, Paris, France
- Cell Biology and Metabolomics platforms, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
46
|
Ghione S, Racoeur C, Mabrouk N, Shan J, Groetz E, Ballot E, Truntzer C, Chouchane L, Végran F, Paul C, Plenchette S, Bettaieb A. Protein Kinase Inhibitor-Mediated Immunoprophylactic and Immunotherapeutic Control of Colon Cancer. Front Immunol 2022; 13:875764. [PMID: 35572581 PMCID: PMC9097540 DOI: 10.3389/fimmu.2022.875764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/24/2022] [Indexed: 12/24/2022] Open
Abstract
Immunotherapy has allowed major advances in oncology in the past years, in particular with the development of immune checkpoint inhibitors, but the clinical benefits are still limited, particularly in colorectal cancer (CRC). Our scientific approach is based on the search for innovative immunotherapy with a final goal that aims to induce an effective antitumor immune response in CRC. Here, we focused on a multikinase inhibitor, H89. We carried out in vivo experiments based on syngeneic mouse models of colon cancer in BALB/c mice and chemically colon tumorigenesis. Flow cytometry, RNAseq, RT-qPCR, antibody-specific immune cell depletion, and Western blot were used to identify the immune cell type involved in the preventive and antitumor activity of H89. We demonstrated that H89 delays colon oncogenesis and prevents tumor growth. This latter effect seems to involve NK cells. H89 also inhibits colon tumor growth in a T-cell-dependent manner. Analysis of the immune landscape in the tumor microenvironment showed an increase of CD4+ Th1 cells and CD8+ cytotoxic T cells but a decrease of CD4+ Treg cell infiltration. Mechanistically, we showed that H89 could promote naïve CD4+ T-cell differentiation into Th1, a decrease in Treg differentiation, and an increase in CD8+ T-cell activation and cytotoxicity ex vivo. Furthermore, H89 induced overexpression of genes involved in antitumor immune response, such as IL-15RA, which depletion counteracts the antitumor effect of H89. We also found that H89 regulated Akt/PP2A pathway axis, involved in TCR and IL-15 signaling transduction. Our findings identify the H89 as a potential strategy for immune system activation leading to the prevention and treatment of CRC.
Collapse
Affiliation(s)
- Silvia Ghione
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC), EA7269, Université Bourgogne Franche-Comté, Dijon, France.,LIIC, Ecole Pratique des Hautes Etudes (EPHE), Paris Sciences et Lettres (PSL) Research University, Paris, France
| | - Cindy Racoeur
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC), EA7269, Université Bourgogne Franche-Comté, Dijon, France.,LIIC, Ecole Pratique des Hautes Etudes (EPHE), Paris Sciences et Lettres (PSL) Research University, Paris, France
| | - Nesrine Mabrouk
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC), EA7269, Université Bourgogne Franche-Comté, Dijon, France.,LIIC, Ecole Pratique des Hautes Etudes (EPHE), Paris Sciences et Lettres (PSL) Research University, Paris, France
| | - Jingxuan Shan
- Genetic Intelligence Laboratory, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar
| | - Emma Groetz
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC), EA7269, Université Bourgogne Franche-Comté, Dijon, France.,LIIC, Ecole Pratique des Hautes Etudes (EPHE), Paris Sciences et Lettres (PSL) Research University, Paris, France
| | - Elise Ballot
- Plateforme de Transfert en Biologie Cancérologique, Centre Georges François Leclerc, Dijon, France.,Team CAdIR, Institut National de la Santé et de la Recherche Médicale (INSERM) U1231, Lipids, Nutrition and Cancer, Dijon, France
| | - Caroline Truntzer
- Plateforme de Transfert en Biologie Cancérologique, Centre Georges François Leclerc, Dijon, France.,Team CAdIR, Institut National de la Santé et de la Recherche Médicale (INSERM) U1231, Lipids, Nutrition and Cancer, Dijon, France
| | - Lotfi Chouchane
- Genetic Intelligence Laboratory, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar
| | - Frédérique Végran
- Plateforme de Transfert en Biologie Cancérologique, Centre Georges François Leclerc, Dijon, France.,Team CAdIR, Institut National de la Santé et de la Recherche Médicale (INSERM) U1231, Lipids, Nutrition and Cancer, Dijon, France.,University of Burgundy and Franche-Comté, Dijon, France
| | - Catherine Paul
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC), EA7269, Université Bourgogne Franche-Comté, Dijon, France.,LIIC, Ecole Pratique des Hautes Etudes (EPHE), Paris Sciences et Lettres (PSL) Research University, Paris, France
| | - Stéphanie Plenchette
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC), EA7269, Université Bourgogne Franche-Comté, Dijon, France.,LIIC, Ecole Pratique des Hautes Etudes (EPHE), Paris Sciences et Lettres (PSL) Research University, Paris, France
| | - Ali Bettaieb
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC), EA7269, Université Bourgogne Franche-Comté, Dijon, France.,LIIC, Ecole Pratique des Hautes Etudes (EPHE), Paris Sciences et Lettres (PSL) Research University, Paris, France
| |
Collapse
|
47
|
Abdellatif M, Bugger H, Kroemer G, Sedej S. NAD + and Vascular Dysfunction: From Mechanisms to Therapeutic Opportunities. J Lipid Atheroscler 2022; 11:111-132. [PMID: 35656147 PMCID: PMC9133775 DOI: 10.12997/jla.2022.11.2.111] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 11/09/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential and pleiotropic coenzyme involved not only in cellular energy metabolism, but also in cell signaling, epigenetic regulation, and post-translational protein modifications. Vascular disease risk factors are associated with aberrant NAD+ metabolism. Conversely, the therapeutic increase of NAD+ levels through the administration of NAD+ precursors or inhibitors of NAD+-consuming enzymes reduces chronic low-grade inflammation, reactivates autophagy and mitochondrial biogenesis, and enhances oxidative metabolism in vascular cells of humans and rodents with vascular pathologies. As such, NAD+ has emerged as a potential target for combatting age-related cardiovascular and cerebrovascular disorders. This review discusses NAD+-regulated mechanisms critical for vascular health and summarizes new advances in NAD+ research directly related to vascular aging and disease, including hypertension, atherosclerosis, coronary artery disease, and aortic aneurysms. Finally, we enumerate challenges and opportunities for NAD+ repletion therapy while anticipating the future of this exciting research field, which will have a major impact on vascular medicine.
Collapse
Affiliation(s)
- Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, Graz, Austria
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
| | - Heiko Bugger
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| |
Collapse
|
48
|
Targeting the gut and tumor microbiota in cancer. Nat Med 2022; 28:690-703. [PMID: 35440726 DOI: 10.1038/s41591-022-01779-2] [Citation(s) in RCA: 274] [Impact Index Per Article: 91.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/09/2022] [Indexed: 02/07/2023]
Abstract
Microorganisms within the gut and other niches may contribute to carcinogenesis, as well as shaping cancer immunosurveillance and response to immunotherapy. Our understanding of the complex relationship between different host-intrinsic microorganisms, as well as the multifaceted mechanisms by which they influence health and disease, has grown tremendously-hastening development of novel therapeutic strategies that target the microbiota to improve treatment outcomes in cancer. Accordingly, the evaluation of a patient's microbial composition and function and its subsequent targeted modulation represent key elements of future multidisciplinary and precision-medicine approaches. In this Review, we outline the current state of research toward harnessing the microbiome to better prevent and treat cancer.
Collapse
|
49
|
Fucikova J, Palova-Jelinkova L, Klapp V, Holicek P, Lanickova T, Kasikova L, Drozenova J, Cibula D, Álvarez-Abril B, García-Martínez E, Spisek R, Galluzzi L. Immunological control of ovarian carcinoma by chemotherapy and targeted anticancer agents. Trends Cancer 2022; 8:426-444. [PMID: 35181272 DOI: 10.1016/j.trecan.2022.01.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/05/2022] [Accepted: 01/19/2022] [Indexed: 12/24/2022]
Abstract
At odds with other solid tumors, epithelial ovarian cancer (EOC) is poorly sensitive to immune checkpoint inhibitors (ICIs), largely reflecting active immunosuppression despite CD8+ T cell infiltration at baseline. Accumulating evidence indicates that both conventional chemotherapeutics and targeted anticancer agents commonly used in the clinical management of EOC not only mediate a cytostatic and cytotoxic activity against malignant cells, but also drive therapeutically relevant immunostimulatory or immunosuppressive effects. Here, we discuss such an immunomodulatory activity, with a specific focus on molecular and cellular pathways that can be harnessed to develop superior combinatorial regimens for clinical EOC care.
Collapse
Affiliation(s)
- Jitka Fucikova
- Sotio, Prague, Czech Republic; Department of Immunology, Charles University, Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic.
| | - Lenka Palova-Jelinkova
- Department of Immunology, Charles University, Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Vanessa Klapp
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Peter Holicek
- Sotio, Prague, Czech Republic; Department of Immunology, Charles University, Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Tereza Lanickova
- Sotio, Prague, Czech Republic; Department of Immunology, Charles University, Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | | | - Jana Drozenova
- Department of Pathology, Third Faculty of Medicine and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - David Cibula
- Gynecologic Oncology Center, Department of Obstetrics and Gynecology, Charles University, First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Beatriz Álvarez-Abril
- Department of Hematology and Oncology, Hospital Universitario Morales Meseguer, Murcia, Spain
| | - Elena García-Martínez
- Department of Hematology and Oncology, Hospital Universitario Morales Meseguer, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain; Universidad Católica San Antonio de Murcia, Guadalupe, Spain
| | - Radek Spisek
- Sotio, Prague, Czech Republic; Department of Immunology, Charles University, Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Centre, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
50
|
Kroemer G, Galassi C, Zitvogel L, Galluzzi L. Immunogenic cell stress and death. Nat Immunol 2022; 23:487-500. [PMID: 35145297 DOI: 10.1038/s41590-022-01132-2] [Citation(s) in RCA: 688] [Impact Index Per Article: 229.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/07/2022] [Indexed: 12/20/2022]
Abstract
Dying mammalian cells emit numerous signals that interact with the host to dictate the immunological correlates of cellular stress and death. In the absence of reactive antigenic determinants (which is generally the case for healthy cells), such signals may drive inflammation but cannot engage adaptive immunity. Conversely, when cells exhibit sufficient antigenicity, as in the case of infected or malignant cells, their death can culminate with adaptive immune responses that are executed by cytotoxic T lymphocytes and elicit immunological memory. Suggesting a key role for immunogenic cell death (ICD) in immunosurveillance, both pathogens and cancer cells evolved strategies to prevent the recognition of cell death as immunogenic. Intriguingly, normal cells succumbing to conditions that promote the formation of post-translational neoantigens (for example, oxidative stress) can also drive at least some degree of antigen-specific immunity, pointing to a novel implication of ICD in the etiology of non-infectious, non-malignant disorders linked to autoreactivity.
Collapse
Affiliation(s)
- Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, Centre de Recherche des Cordeliers, INSERM U1138, Sorbonne Université, Université de Paris, Institut Universitaire de France, Paris, France. .,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France. .,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Claudia Galassi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Université Paris Saclay, Faculty of Medicine, Le Kremlin-Bicêtre, France.,INSERM U1015, Villejuif, France.,Equipe labellisée par la Ligue contre le cancer, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) BIOTHERIS, Villejuif, France
| | - 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
|