51
|
Battaglia TW, Mimpen IL, Traets JJH, van Hoeck A, Zeverijn LJ, Geurts BS, de Wit GF, Noë M, Hofland I, Vos JL, Cornelissen S, Alkemade M, Broeks A, Zuur CL, Cuppen E, Wessels L, van de Haar J, Voest E. A pan-cancer analysis of the microbiome in metastatic cancer. Cell 2024; 187:2324-2335.e19. [PMID: 38599211 DOI: 10.1016/j.cell.2024.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/30/2023] [Accepted: 03/18/2024] [Indexed: 04/12/2024]
Abstract
Microbial communities are resident to multiple niches of the human body and are important modulators of the host immune system and responses to anticancer therapies. Recent studies have shown that complex microbial communities are present within primary tumors. To investigate the presence and relevance of the microbiome in metastases, we integrated mapping and assembly-based metagenomics, genomics, transcriptomics, and clinical data of 4,160 metastatic tumor biopsies. We identified organ-specific tropisms of microbes, enrichments of anaerobic bacteria in hypoxic tumors, associations between microbial diversity and tumor-infiltrating neutrophils, and the association of Fusobacterium with resistance to immune checkpoint blockade (ICB) in lung cancer. Furthermore, longitudinal tumor sampling revealed temporal evolution of the microbial communities and identified bacteria depleted upon ICB. Together, we generated a pan-cancer resource of the metastatic tumor microbiome that may contribute to advancing treatment strategies.
Collapse
Affiliation(s)
- Thomas W Battaglia
- Division of Molecular Oncology & Immunology, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands; Division of Molecular Carcinogenesis, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands; Oncode Institute, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Iris L Mimpen
- Division of Molecular Oncology & Immunology, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands; Oncode Institute, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Joleen J H Traets
- Division of Tumor Biology & Immunology, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Arne van Hoeck
- Oncode Institute, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands; Department of Head and Neck Surgery and Oncology, the Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Laurien J Zeverijn
- Division of Molecular Oncology & Immunology, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands; Oncode Institute, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Birgit S Geurts
- Division of Molecular Oncology & Immunology, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands; Oncode Institute, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Gijs F de Wit
- Division of Molecular Oncology & Immunology, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands; Oncode Institute, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Michaël Noë
- Department of Pathology, Antoni van Leeuwenhoek/the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ingrid Hofland
- Core Facility Molecular Pathology & Biobanking, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Joris L Vos
- Division of Tumor Biology & Immunology, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands; Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sten Cornelissen
- Core Facility Molecular Pathology & Biobanking, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Maartje Alkemade
- Core Facility Molecular Pathology & Biobanking, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Annegien Broeks
- Core Facility Molecular Pathology & Biobanking, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Charlotte L Zuur
- Division of Tumor Biology & Immunology, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands; Department of Head and Neck Surgery and Oncology, the Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Department of Otorhinolaryngology Head and Neck Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Edwin Cuppen
- Center for Molecular Medicine, University Medical Centre Utrecht, Utrecht 3584CX, the Netherlands; Hartwig Medical Foundation, Science Park, Amsterdam 1098XH, the Netherlands
| | - Lodewyk Wessels
- Division of Molecular Carcinogenesis, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands; Oncode Institute, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands; Faculty of EEMCS, Delft University of Technology, Delft 2628 CD, the Netherlands
| | - Joris van de Haar
- Division of Molecular Oncology & Immunology, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands; Oncode Institute, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Emile Voest
- Division of Molecular Oncology & Immunology, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands; Oncode Institute, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands.
| |
Collapse
|
52
|
Knoedler L, Huelsboemer L, Hollmann K, Alfertshofer M, Herfeld K, Hosseini H, Boroumand S, Stoegner VA, Safi AF, Perl M, Knoedler S, Pomahac B, Kauke-Navarro M. From standard therapies to monoclonal antibodies and immune checkpoint inhibitors - an update for reconstructive surgeons on common oncological cases. Front Immunol 2024; 15:1276306. [PMID: 38715609 PMCID: PMC11074450 DOI: 10.3389/fimmu.2024.1276306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 04/05/2024] [Indexed: 05/23/2024] Open
Abstract
Malignancies represent a persisting worldwide health burden. Tumor treatment is commonly based on surgical and/or non-surgical therapies. In the recent decade, novel non-surgical treatment strategies involving monoclonal antibodies (mAB) and immune checkpoint inhibitors (ICI) have been successfully incorporated into standard treatment algorithms. Such emerging therapy concepts have demonstrated improved complete remission rates and prolonged progression-free survival compared to conventional chemotherapies. However, the in-toto surgical tumor resection followed by reconstructive surgery oftentimes remains the only curative therapy. Breast cancer (BC), skin cancer (SC), head and neck cancer (HNC), and sarcoma amongst other cancer entities commonly require reconstructive surgery to restore form, aesthetics, and functionality. Understanding the basic principles, strengths, and limitations of mAB and ICI as (neo-) adjuvant therapies and treatment alternatives for resectable or unresectable tumors is paramount for optimized surgical therapy planning. Yet, there is a scarcity of studies that condense the current body of literature on mAB and ICI for BC, SC, HNC, and sarcoma. This knowledge gap may result in suboptimal treatment planning, ultimately impairing patient outcomes. Herein, we aim to summarize the current translational endeavors focusing on mAB and ICI. This line of research may serve as an evidence-based fundament to guide targeted therapy and optimize interdisciplinary anti-cancer strategies.
Collapse
Affiliation(s)
- Leonard Knoedler
- Department of Plastic, Hand, and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Lioba Huelsboemer
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Katharina Hollmann
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Faculty of Medicine, University of Wuerzbuerg, Wuerzburg, Germany
| | - Michael Alfertshofer
- Division of Hand, Plastic and Aesthetic Surgery, Ludwig-Maximilians University Munich, Munich, Germany
| | - Konstantin Herfeld
- Department of Internal Medicine III (Oncology and Haematology), University Hospital Regensburg, Regensburg, Germany
- Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Helia Hosseini
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Sam Boroumand
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Viola A. Stoegner
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Burn Center, Hannover Medical School, Hannover, Germany
| | - Ali-Farid Safi
- Craniologicum, Center for Cranio-Maxillo-Facial Surgery, Bern, Switzerland
- Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Markus Perl
- Department of Internal Medicine III (Oncology and Haematology), University Hospital Regensburg, Regensburg, Germany
- Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Samuel Knoedler
- Department of Plastic, Hand, and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Bohdan Pomahac
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Martin Kauke-Navarro
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
53
|
Lei X, de Groot DC, Welters MJP, de Wit T, Schrama E, van Eenennaam H, Santegoets SJ, Oosenbrug T, van der Veen A, Vos JL, Zuur CL, de Miranda NFCC, Jacobs H, van der Burg SH, Borst J, Xiao Y. CD4 + T cells produce IFN-I to license cDC1s for induction of cytotoxic T-cell activity in human tumors. Cell Mol Immunol 2024; 21:374-392. [PMID: 38383773 PMCID: PMC10978876 DOI: 10.1038/s41423-024-01133-1] [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: 07/11/2023] [Accepted: 01/05/2024] [Indexed: 02/23/2024] Open
Abstract
CD4+ T cells can "help" or "license" conventional type 1 dendritic cells (cDC1s) to induce CD8+ cytotoxic T lymphocyte (CTL) anticancer responses, as proven in mouse models. We recently identified cDC1s with a transcriptomic imprint of CD4+ T-cell help, specifically in T-cell-infiltrated human cancers, and these cells were associated with a good prognosis and response to PD-1-targeting immunotherapy. Here, we delineate the mechanism of cDC1 licensing by CD4+ T cells in humans. Activated CD4+ T cells produce IFNβ via the STING pathway, which promotes MHC-I antigen (cross-)presentation by cDC1s and thereby improves their ability to induce CTL anticancer responses. In cooperation with CD40 ligand (L), IFNβ also optimizes the costimulatory and other functions of cDC1s required for CTL response induction. IFN-I-producing CD4+ T cells are present in diverse T-cell-infiltrated cancers and likely deliver "help" signals to CTLs locally, according to their transcriptomic profile and colocalization with "helped/licensed" cDCs and tumor-reactive CD8+ T cells. In agreement with this scenario, the presence of IFN-I-producing CD4+ T cells in the TME is associated with overall survival and the response to PD-1 checkpoint blockade in cancer patients.
Collapse
Affiliation(s)
- Xin Lei
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
- Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Daniël C de Groot
- Department of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marij J P Welters
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom de Wit
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
- Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Ellen Schrama
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
- Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Saskia J Santegoets
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Timo Oosenbrug
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Joris L Vos
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Charlotte L Zuur
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Otorhinolaryngology Leiden University Medical Center, Leiden, The Netherlands
| | | | - Heinz Jacobs
- Department of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sjoerd H van der Burg
- Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Jannie Borst
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands.
- Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands.
| | - Yanling Xiao
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands.
- Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
54
|
Vaidya P, Cohen EE. Facts and Hopes in Neoadjuvant Immunotherapy: Current Approvals and Emerging Evidence. Clin Cancer Res 2024; 30:1232-1239. [PMID: 37955563 PMCID: PMC10984792 DOI: 10.1158/1078-0432.ccr-23-0583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/27/2023] [Accepted: 11/09/2023] [Indexed: 11/14/2023]
Abstract
In 2021 and 2022, two immune checkpoint inhibitors received FDA approval in the neoadjuvant setting for the treatment of early-stage triple negative breast cancer (TNBC) and non-small cell lung cancer (NSCLC). Several more studies have since indicated the benefits, and challenges, of administering neoadjuvant immunotherapy prior to definitive surgery in the gastrointestinal, head and neck, and cutaneous realms. In addition, numerous ongoing phase II and phase III trials are investigating outcomes of neoadjuvant immune treatment in early-stage disease. As such, it is anticipated that more immune checkpoint inhibitors will receive approval for various neoadjuvant indications in the next several years. Medical oncologists, surgeons, and other providers in a multidisciplinary cancer care team will be presented with alternate treatment paradigms and clinical decisions regarding upfront surgery versus neoadjuvant treatment. Here, we describe the current evidence supporting use of immune checkpoint inhibitors for neoadjuvant treatment, ongoing studies, and clinical considerations of this treatment approach.
Collapse
Affiliation(s)
- Poorva Vaidya
- Dept of Internal Medicine, Division of Hematology-Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Ezra E.W. Cohen
- Dept of Internal Medicine, Division of Hematology-Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA
| |
Collapse
|
55
|
Chen Z, Wang X, Jin Z, Li B, Jiang D, Wang Y, Jiang M, Zhang D, Yuan P, Zhao Y, Feng F, Lin Y, Jiang L, Wang C, Meng W, Ye W, Wang J, Qiu W, Liu H, Huang D, Hou Y, Wang X, Jiao Y, Ying J, Liu Z, Liu Y. Deep learning on tertiary lymphoid structures in hematoxylin-eosin predicts cancer prognosis and immunotherapy response. NPJ Precis Oncol 2024; 8:73. [PMID: 38519580 PMCID: PMC10959936 DOI: 10.1038/s41698-024-00579-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/14/2024] [Indexed: 03/25/2024] Open
Abstract
Tertiary lymphoid structures (TLSs) have been associated with favorable immunotherapy responses and prognosis in various cancers. Despite their significance, their quantification using multiplex immunohistochemistry (mIHC) staining of T and B lymphocytes remains labor-intensive, limiting its clinical utility. To address this challenge, we curated a dataset from matched mIHC and H&E whole-slide images (WSIs) and developed a deep learning model for automated segmentation of TLSs. The model achieved Dice coefficients of 0.91 on the internal test set and 0.866 on the external validation set, along with intersection over union (IoU) scores of 0.819 and 0.787, respectively. The TLS ratio, defined as the segmented TLS area over the total tissue area, correlated with B lymphocyte levels and the expression of CXCL13, a chemokine associated with TLS formation, in 6140 patients spanning 16 tumor types from The Cancer Genome Atlas (TCGA). The prognostic models for overall survival indicated that the inclusion of the TLS ratio with TNM staging significantly enhanced the models' discriminative ability, outperforming the traditional models that solely incorporated TNM staging, in 10 out of 15 TCGA tumor types. Furthermore, when applied to biopsied treatment-naïve tumor samples, higher TLS ratios predicted a positive immunotherapy response across multiple cohorts, including specific therapies for esophageal squamous cell carcinoma, non-small cell lung cancer, and stomach adenocarcinoma. In conclusion, our deep learning-based approach offers an automated and reproducible method for TLS segmentation and quantification, highlighting its potential in predicting immunotherapy response and informing cancer prognosis.
Collapse
Affiliation(s)
- Ziqiang Chen
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xiaobing Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zelin Jin
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Bosen Li
- Department of General Surgery/Gastric Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dongxian Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanqiu Wang
- Departments of Pathology, International Peace Maternity and Child Health Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengping Jiang
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Dandan Zhang
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Pei Yuan
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yahui Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Feiyue Feng
- Thoracic Surgery Department, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yicheng Lin
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Liping Jiang
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chenxi Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weida Meng
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Wenjing Ye
- Division of Rheumatology and Immunology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jie Wang
- Departments of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wenqing Qiu
- Shanghai Xuhui Central Hospital, Shanghai, China
| | - Houbao Liu
- Shanghai Xuhui Central Hospital, Shanghai, China
- Department of General Surgery/Biliary Tract Disease Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dan Huang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Institute of Pathology, Fudan University, Shanghai, China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xuefei Wang
- Department of General Surgery/Gastric Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital (Xiamen), Fudan University, Shanghai, China
| | - Yuchen Jiao
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yun Liu
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
56
|
Gil-Jimenez A, van Dijk N, Vos JL, Lubeck Y, van Montfoort ML, Peters D, Hooijberg E, Broeks A, Zuur CL, van Rhijn BWG, Vis DJ, van der Heijden MS, Wessels LFA. Spatial relationships in the urothelial and head and neck tumor microenvironment predict response to combination immune checkpoint inhibitors. Nat Commun 2024; 15:2538. [PMID: 38514623 PMCID: PMC10957922 DOI: 10.1038/s41467-024-46450-1] [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: 05/09/2023] [Accepted: 02/28/2024] [Indexed: 03/23/2024] Open
Abstract
Immune checkpoint inhibitors (ICI) can achieve remarkable responses in urothelial cancer (UC), which may depend on tumor microenvironment (TME) characteristics. However, the relationship between the TME, usually characterized by immune cell density, and response to ICI is unclear. Here, we quantify the TME immune cell densities and spatial relationships (SRs) of 24 baseline UC samples, obtained before pre-operative combination ICI treatment, using multiplex immunofluorescence. We describe SRs by approximating the first nearest-neighbor distance distribution with a Weibull distribution and evaluate the association between TME metrics and ipilimumab+nivolumab response. Immune cell density does not discriminate between response groups. However, the Weibull SR metrics of CD8+ T cells or macrophages to their closest cancer cell positively associate with response. CD8+ T cells close to B cells are characteristic of non-response. We validate our SR response associations in a combination ICI cohort of head and neck tumors. Our data confirm that SRs, in contrast to density metrics, are strong biomarkers of response to pre-operative combination ICIs.
Collapse
Affiliation(s)
- Alberto Gil-Jimenez
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Nick van Dijk
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Joris L Vos
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Yoni Lubeck
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Dennis Peters
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Erik Hooijberg
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Annegien Broeks
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Charlotte L Zuur
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Otorhinolaryngology Head and Neck Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Bas W G van Rhijn
- Department of Urology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Urology, Caritas St. Josef Medical Centre, University of Regensburg, Regensburg, Germany
| | - Daniel J Vis
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Michiel S van der Heijden
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Lodewyk F A Wessels
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
- Oncode Institute, Utrecht, the Netherlands.
- Faculty of Electrical Engineering, Mathematics and Computer Science, Delft University of Technology, Delft, the Netherlands.
| |
Collapse
|
57
|
Franken A, Bila M, Mechels A, Kint S, Van Dessel J, Pomella V, Vanuytven S, Philips G, Bricard O, Xiong J, Boeckx B, Hatse S, Van Brussel T, Schepers R, Van Aerde C, Geurs S, Vandecaveye V, Hauben E, Vander Poorten V, Verbandt S, Vandereyken K, Qian J, Tejpar S, Voet T, Clement PM, Lambrechts D. CD4 + T cell activation distinguishes response to anti-PD-L1+anti-CTLA4 therapy from anti-PD-L1 monotherapy. Immunity 2024; 57:541-558.e7. [PMID: 38442708 DOI: 10.1016/j.immuni.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 11/30/2023] [Accepted: 02/08/2024] [Indexed: 03/07/2024]
Abstract
Cancer patients often receive a combination of antibodies targeting programmed death-ligand 1 (PD-L1) and cytotoxic T lymphocyte antigen-4 (CTLA4). We conducted a window-of-opportunity study in head and neck squamous cell carcinoma (HNSCC) to examine the contribution of anti-CTLA4 to anti-PD-L1 therapy. Single-cell profiling of on- versus pre-treatment biopsies identified T cell expansion as an early response marker. In tumors, anti-PD-L1 triggered the expansion of mostly CD8+ T cells, whereas combination therapy expanded both CD4+ and CD8+ T cells. Such CD4+ T cells exhibited an activated T helper 1 (Th1) phenotype. CD4+ and CD8+ T cells co-localized with and were surrounded by dendritic cells expressing T cell homing factors or antibody-producing plasma cells. T cell receptor tracing suggests that anti-CTLA4, but not anti-PD-L1, triggers the trafficking of CD4+ naive/central-memory T cells from tumor-draining lymph nodes (tdLNs), via blood, to the tumor wherein T cells acquire a Th1 phenotype. Thus, CD4+ T cell activation and recruitment from tdLNs are hallmarks of early response to anti-PD-L1 plus anti-CTLA4 in HNSCC.
Collapse
Affiliation(s)
- Amelie Franken
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; VIB Center for Cancer Biology, Leuven 3000, Belgium
| | - Michel Bila
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, 3000 Leuven, Belgium; Department of General Medical Oncology, UZ Leuven, 3000 Leuven, Belgium; Department of Oral and Maxillofacial Surgery, UZ Leuven, Leuven 3000, Belgium
| | - Aurelie Mechels
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; VIB Center for Cancer Biology, Leuven 3000, Belgium
| | - Sam Kint
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; KU Leuven Institute for Single Cell Omics (LISCO), Leuven 3000, Belgium
| | - Jeroen Van Dessel
- Department of Oral and Maxillofacial Surgery, UZ Leuven, Leuven 3000, Belgium
| | | | - Sebastiaan Vanuytven
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; KU Leuven Institute for Single Cell Omics (LISCO), Leuven 3000, Belgium
| | - Gino Philips
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; VIB Center for Cancer Biology, Leuven 3000, Belgium
| | - Orian Bricard
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; VIB Center for Cancer Biology, Leuven 3000, Belgium
| | - Jieyi Xiong
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; VIB Center for Cancer Biology, Leuven 3000, Belgium
| | - Bram Boeckx
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; VIB Center for Cancer Biology, Leuven 3000, Belgium
| | - Sigrid Hatse
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, 3000 Leuven, Belgium; Department of General Medical Oncology, UZ Leuven, 3000 Leuven, Belgium
| | - Thomas Van Brussel
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; VIB Center for Cancer Biology, Leuven 3000, Belgium
| | - Rogier Schepers
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; VIB Center for Cancer Biology, Leuven 3000, Belgium
| | - Cedric Van Aerde
- Department of Imaging and Pathology, KU Leuven, UZ Leuven, Leuven 3000, Belgium
| | - Sarah Geurs
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; KU Leuven Institute for Single Cell Omics (LISCO), Leuven 3000, Belgium; Department of Biomolecular Medicine, UZ Ghent, Ghent 9052, Belgium
| | | | - Esther Hauben
- Otorhinolaryngology, Head and Neck Surgery, Leuven 3000, Belgium
| | - Vincent Vander Poorten
- Otorhinolaryngology, Head and Neck Surgery, Leuven 3000, Belgium; Department of Oncology, Section Head and Neck Oncology, Leuven 3000, Belgium
| | - Sara Verbandt
- Digestive Oncology, KU Leuven, UZ Leuven, Leuven 3000, Belgium
| | - Katy Vandereyken
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; KU Leuven Institute for Single Cell Omics (LISCO), Leuven 3000, Belgium
| | - Junbin Qian
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Institute of Genetics, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Sabine Tejpar
- Digestive Oncology, KU Leuven, UZ Leuven, Leuven 3000, Belgium
| | - Thierry Voet
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; KU Leuven Institute for Single Cell Omics (LISCO), Leuven 3000, Belgium
| | - Paul M Clement
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, 3000 Leuven, Belgium; Department of General Medical Oncology, UZ Leuven, 3000 Leuven, Belgium.
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; VIB Center for Cancer Biology, Leuven 3000, Belgium; KU Leuven Institute for Single Cell Omics (LISCO), Leuven 3000, Belgium.
| |
Collapse
|
58
|
Wu D, Li Y, Xu P, Fang Q, Cao F, Lin H, Li Y, Su Y, Lu L, Chen L, Li Y, Zhao Z, Hong X, Li G, Tian Y, Sun J, Yan H, Fan Y, Zhang X, Li Z, Liu X. Neoadjuvant chemo-immunotherapy with camrelizumab plus nab-paclitaxel and cisplatin in resectable locally advanced squamous cell carcinoma of the head and neck: a pilot phase II trial. Nat Commun 2024; 15:2177. [PMID: 38467604 PMCID: PMC10928200 DOI: 10.1038/s41467-024-46444-z] [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: 07/18/2023] [Accepted: 02/28/2024] [Indexed: 03/13/2024] Open
Abstract
Neoadjuvant chemoimmunotherapy has emerged as a potential treatment option for resectable head and neck squamous cell carcinoma (HNSCC). In this single-arm phase II trial (NCT04826679), patients with resectable locally advanced HNSCC (T2‒T4, N0‒N3b, M0) received neoadjuvant chemoimmunotherapy with camrelizumab (200 mg), nab-paclitaxel (260 mg/m2), and cisplatin (60 mg/m2) intravenously on day one of each three-week cycle for three cycles. The primary endpoint was the objective response rate (ORR). Secondary endpoints included pathologic complete response (pCR), major pathologic response (MPR), two-year progression-free survival rate, two-year overall survival rate, and toxicities. Here, we report the perioperative outcomes; survival outcomes were not mature at the time of data analysis. Between April 19, 2021 and March 17, 2022, 48 patients were enrolled and received neoadjuvant therapy, 27 of whom proceeded to surgical resection and remaining 21 received non-surgical therapy. The ORR was 89.6% (95% CI: 80.9, 98.2) among 48 patients who completed neoadjuvant therapy. Of the 27 patients who underwent surgery, 17 (63.0%, 95% CI: 44.7, 81.2) achieved a MPR or pCR, with a pCR rate of 55.6% (95% CI: 36.8, 74.3). Treatment-related adverse events of grade 3 or 4 occurred in two patients. This study meets the primary endpoint showing potential efficacy of neoadjuvant camrelizumab plus nab-paclitaxel and cisplatin, with an acceptable safety profile, in patients with resectable locally advanced HNSCC.
Collapse
Affiliation(s)
- Di Wu
- Department of Head and Neck Surgery, Sun Yat-Sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yong Li
- Department of Pathology, Sun Yat-Sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Pengfei Xu
- Department of Head and Neck Surgery, Sun Yat-Sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Qi Fang
- Department of Head and Neck Surgery, Sun Yat-Sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Fei Cao
- Department of Head and Neck Surgery, Sun Yat-Sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Hongsheng Lin
- Department of Head and Neck Surgery, Sun Yat-Sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yin Li
- Department of Head and Neck Surgery, Sun Yat-Sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yong Su
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicin, Guangzhou, China
| | - Lixia Lu
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicin, Guangzhou, China
| | - Lei Chen
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicin, Guangzhou, China
| | - Yizhuo Li
- Department of Radiology, Sun Yat-Sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zheng Zhao
- Department of Head and Neck Surgery, Sun Yat-Sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiaoyu Hong
- Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Guohong Li
- Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Yaru Tian
- Jiangsu Hengrui Pharmaceuticals Co., LTD, Shanghai, China
| | - Jinyun Sun
- Jiangsu Hengrui Pharmaceuticals Co., LTD, Shanghai, China
| | - Honghong Yan
- Department of Intensive Care Medicine, Sun Yat-Sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yunyun Fan
- Department of Head and Neck Surgery, Sun Yat-Sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xinrui Zhang
- Department of Otolaryngology-Head and Neck Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Zhiming Li
- Department of Medical Oncology, Sun Yat-Sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Xuekui Liu
- Department of Head and Neck Surgery, Sun Yat-Sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
59
|
Wu SG, Wang RJ, Zhou Y, Luo XY. Neoadjuvant therapy with chemotherapy and immune checkpoint inhibitor for laryngeal function preservation in locally advanced hypopharyngeal cancer. Front Immunol 2024; 15:1364799. [PMID: 38524124 PMCID: PMC10958379 DOI: 10.3389/fimmu.2024.1364799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 02/23/2024] [Indexed: 03/26/2024] Open
Abstract
Purpose To evaluate the efficacy and laryngeal function preservation of neoadjuvant treatment with chemotherapy and immune checkpoint inhibitor for locally advanced hypopharyngeal cancer (LAHPC). Methods We retrospectively collected LAHPC patients who were diagnosed between February 2022 and June 2023. The patients received a combination of chemotherapy and immune checkpoint inhibitors as the neoadjuvant therapy. The response to treatment, laryngeal function preservation rate, and short-term survival were assessed. Results A total of 20 patients were included. Of these patients, 17 (85.0%) had stage IVA-B disease. Ten (50%) and four (20%) patients achieved pathological complete response (PCR) and major pathological response (MPR) to the primary tumor, respectively. In addition, 6 patients had incomplete pathological response (IPR). In the neck, 19 patients had node-positive disease before treatment, and only 5 patients (26.4%) had PCR to regional lymph nodes. Pathologically positive lymph nodes were still observed in 14 (73.6%) patients. Significant downgrading on narrow-band imaging assessment in primary tumors was associated with a higher probability of PCR or MPR than those with IPR (92.9% vs. 33.3%, P=0.014). The overall rate of laryngeal preservation was 95.0%. No severe perioperative complications or perioperative death were found. All patients completed the recommended postoperative radiotherapy/chemoradiotherapy. The median follow-up period was 12.1 months. The 1-year progression-free survival and overall survival were 94.1% and 92.9%, respectively. During the follow-up period, all 19 patients who underwent laryngeal preservation surgery had their laryngeal function preserved. Conclusion The addition of an immune checkpoint inhibitor to neoadjuvant chemotherapy effectively preserves laryngeal function without increasing complications related to surgery and postoperative radiotherapy in LAHPC.
Collapse
Affiliation(s)
- San-Gang Wu
- Department of Radiation Oncology, Xiamen Cancer Quality Control Center, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Run-Jie Wang
- Department of Radiation Oncology, Xiamen Cancer Quality Control Center, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yi Zhou
- Department of Otolaryngology-Head and Neck Surgery, Xiamen Key Laboratory of Otolaryngology-Head and Neck Surgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xian-Yang Luo
- Department of Otolaryngology-Head and Neck Surgery, Xiamen Key Laboratory of Otolaryngology-Head and Neck Surgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
60
|
Chen J, Tang S, Zheng Q, Li J, Jiang H, Lu H, Liao G, Li K, Liang Y. The competitive mechanism of EZH1 and EZH2 in promoting oral squamous cell carcinoma. Exp Cell Res 2024; 436:113957. [PMID: 38309675 DOI: 10.1016/j.yexcr.2024.113957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/23/2024] [Accepted: 01/30/2024] [Indexed: 02/05/2024]
Abstract
Enhancer of Zeste Homolog 1 (EZH1) and Enhancer of Zeste Homolog 2 (EZH2) are the key components of polycomb repressive complex 2 (PRC2); however, the roles of these proteins in oral squamous cell carcinoma (OSCC) have yet to be elucidated. In this study, we aimed to determine the respective roles of these proteins in OSCC by investigating the expression levels of EZH1 and EZH2 in OSCC tissues (N = 63) by immunohistochemistry. In addition, we used lentiviruses to construct stable OSCC cell lines that overexpressed EZH1 and EZH2. Then, we investigated these cell lines for cell viability, colony formation capacity, stemness, and epithelial-mesenchymal transition (EMT). Binding competition between EZH1 and EZH2 with PRC2 was further evaluated using Co-immunoprecipitation (Co-IP). Compared with normal tissues, the expression levels of EZH2 in OSCC tissues was up-regulated, while the expression of EZH1 was down-regulated. EZH2 enhanced cell viability, colony formation capacity, stemness, and EMT, while EZH1 did not. Furthermore, analysis indicated that EZH1 and EZH2 bound competitively to PRC2 and influenced the methylation status of H3K27. In conclusion, our findings verified that EZH1 and EZH2 play opposing roles in OSCC and that EZH1 and EZH2 compete as the key component of PRC2, thus affecting the characteristics of OSCC via the methylation of H3K27.
Collapse
Affiliation(s)
- Jianghai Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling-yuan West Street, Guangzhou, 510000, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Zhong-Shan Er Road 74, Guangzhou, 510080, China
| | - Shanshan Tang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling-yuan West Street, Guangzhou, 510000, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Zhong-Shan Er Road 74, Guangzhou, 510080, China
| | - Qiuhan Zheng
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling-yuan West Street, Guangzhou, 510000, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Zhong-Shan Er Road 74, Guangzhou, 510080, China
| | - Jingyuan Li
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling-yuan West Street, Guangzhou, 510000, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Zhong-Shan Er Road 74, Guangzhou, 510080, China
| | - Hong Jiang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling-yuan West Street, Guangzhou, 510000, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Zhong-Shan Er Road 74, Guangzhou, 510080, China
| | - Huanzi Lu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling-yuan West Street, Guangzhou, 510000, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Zhong-Shan Er Road 74, Guangzhou, 510080, China
| | - Guiqing Liao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling-yuan West Street, Guangzhou, 510000, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Zhong-Shan Er Road 74, Guangzhou, 510080, China.
| | - Kan Li
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling-yuan West Street, Guangzhou, 510000, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Zhong-Shan Er Road 74, Guangzhou, 510080, China.
| | - Yujie Liang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling-yuan West Street, Guangzhou, 510000, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Zhong-Shan Er Road 74, Guangzhou, 510080, China.
| |
Collapse
|
61
|
Hong Q, Ding S, Xing C, Mu Z. Advances in tumor immune microenvironment of head and neck squamous cell carcinoma: A review of literature. Medicine (Baltimore) 2024; 103:e37387. [PMID: 38428879 PMCID: PMC10906580 DOI: 10.1097/md.0000000000037387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/03/2024] Open
Abstract
Squamous cell carcinoma is seen as principal malignancy of head and neck. Tumor immune microenvironment plays a vital role in the occurrence, development and treatment of head and neck squamous cell carcinoma (HNSCC). The effect of immunotherapy, in particular, is closely related to tumor immune microenvironment. This review searched for high-quality literature included within PubMed, Web of Science, and Scopus using the keywords "head and neck cancers," "tumor microenvironment" and "immunotherapy," with the view to summarizing the characteristics of HNSCC immune microenvironment and how various subsets of immune cells promote tumorigenesis. At the same time, based on the favorable prospects of immunotherapy having been shown currently, the study is committed to pinpointing the latest progress of HNSCC immunotherapy, which is of great significance in not only further guiding the diagnosis and treatment of HNSCC, but also conducting its prognostic judgement.
Collapse
Affiliation(s)
- Qichao Hong
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Shun Ding
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Chengliang Xing
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Zhonglin Mu
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| |
Collapse
|
62
|
Feng Y, Guo K, Jin H, Jiang J, Wang M, Lin S. Efficacy and safety of neoadjuvant combination immunotherapy in surgically resectable malignant solid tumors: a systematic review and meta-analysis. Expert Rev Anticancer Ther 2024; 24:169-181. [PMID: 38436076 DOI: 10.1080/14737140.2024.2325404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/18/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVES Neoadjuvant immunotherapy has emerged as a prominent research focus recently. For potentially operable patients, neoadjuvant therapy serves as a primary method to reduce tumor load and facilitate surgical interventions. METHODS We retrieved articles from PubMed, Embase, Cochrane Library, American Society of Clinical Oncology, and European Society of Medical Oncology websites from inception to December 2023. Statistical analyses were performed using the R software. Primary outcomes assessed included major pathological response (MPR), pathological complete response (pCR), and treatment-related adverse events (trAEs). RESULTS 29 studies encompassing 1163 patients were included. The MPR rate of neoadjuvant combination immunotherapy was 38% (95% confidence interval [CI]: 25%-52%), and the pCR rate was 33% (95%CI: 25%-42%). These values were significantly higher than those obtained with single agent immunotherapy (p < 0.001). The pooled incidence of overall trAEs was 83% (95%CI: 73%-92%), and grade (G) 3-4 trAEs was 22% (95%CI: 15%-29%), both significantly higher than those observed with single agent immunotherapy (p < 0.05). CONCLUSION This study demonstrated the efficacy of neoadjuvant combination immunotherapy. Given that the majority of the included trials were phase II with small sample sizes, further multicenter phase III randomized controlled trials should be conducted to validate the findings of the review.
Collapse
Affiliation(s)
- Yuqian Feng
- Hangzhou School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Kaibo Guo
- Department of Oncology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Huimin Jin
- Department of Oncology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jing Jiang
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Menglei Wang
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shengyou Lin
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
63
|
Huang Y, Liu H, Liu B, Chen X, Li D, Xue J, Li N, Zhu L, Yang L, Xiao J, Liu C. Quantified pathway mutations associate epithelial-mesenchymal transition and immune escape with poor prognosis and immunotherapy resistance of head and neck squamous cell carcinoma. BMC Med Genomics 2024; 17:49. [PMID: 38331768 PMCID: PMC10854145 DOI: 10.1186/s12920-024-01818-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Pathway mutations have been calculated to predict the poor prognosis and immunotherapy resistance in head and neck squamous cell carcinoma (HNSCC). To uncover the unique markers predicting prognosis and immune therapy response, the accurate quantification of pathway mutations are required to evaluate epithelial-mesenchymal transition (EMT) and immune escape. Yet, there is a lack of score to accurately quantify pathway mutations. MATERIAL AND METHODS Firstly, we proposed Individualized Weighted Hallmark Gene Set Mutation Burden (IWHMB, https://github.com/YuHongHuang-lab/IWHMB ) which integrated pathway structure information and eliminated the interference of global Tumor Mutation Burden to accurately quantify pathway mutations. Subsequently, to further elucidate the association of IWHMB with EMT and immune escape, support vector machine regression model was used to identify IWHMB-related transcriptomic features (IRG), while Adversarially Regularized Graph Autoencoder (ARVGA) was used to further resolve IRG network features. Finally, Random walk with restart algorithm was used to identify biomarkers for predicting ICI response. RESULTS We quantified the HNSCC pathway mutation signatures and identified pathway mutation subtypes using IWHMB. The IWHMB-related transcriptomic features (IRG) identified by support vector machine regression were divided into 5 communities by ARVGA, among which the Community 1 enriching malignant mesenchymal components promoted EMT dynamically and regulated immune patterns associated with ICI responses. Bridge Hub Gene (BHG) identified by random walk with restart was key to IWHMB in EMT and immune escape, thus, more predictive for ICI response than other 70 public signatures. CONCLUSION In summary, the novel pathway mutation scoring-IWHMB suggested that the elevated malignancy mediated by pathway mutations is a major cause of poor prognosis and immunotherapy failure in HNSCC, and is capable of identifying novel biomarkers to predict immunotherapy response.
Collapse
Affiliation(s)
- Yuhong Huang
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
- Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Han Liu
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
- Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Bo Liu
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, China
| | - Xiaoyan Chen
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
| | - Danya Li
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
| | - Junyuan Xue
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
| | - Nan Li
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
- Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Lei Zhu
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
- Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Liu Yang
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
| | - Jing Xiao
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China.
- Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China.
| | - Chao Liu
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China.
- Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China.
| |
Collapse
|
64
|
Di Giacomo AM, Lahn M, Eggermont AM, Fox B, Ibrahim R, Sharma P, Allison JP, Maio M. The future of targeting cytotoxic T-lymphocyte-associated protein-4: Is there a role? Eur J Cancer 2024; 198:113501. [PMID: 38169219 DOI: 10.1016/j.ejca.2023.113501] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024]
Abstract
The 2022 yearly Think Tank Meeting in Siena, Tuscany (Italy), organized by the Italian Network for Tumor Biotherapy (NIBIT) Foundation, the Parker Institute for Cancer Immunotherapy and the World Immunotherapy Council, included a focus on the future of integrating and expanding the use of targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). The conference members exchanged their views on the lessons from targeting CTLA-4 and compared the effect to the impact of blocking Programmed cell death protein 1 (PD1) or its ligand (PDL1). The increasing experience with both therapeutic approaches and their combination suggests that targeting CTLA-4 may lead to more durable responses for a sizeable proportion of patients, though the specific mechanism is not entirely understood. Overcoming toxicity of blocking CTLA-4 is currently being addressed with different doses and dose regimens, especially when combined with PD1/PDL1 blocking antibodies. Novel therapeutics targeting CTLA-4 hold the promise to reduce toxicities and thus allow different combination strategies in the future. On the whole, the consent was that targeting CTLA-4 remains an important strategy to improve the efficacy of cancer immunotherapies.
Collapse
Affiliation(s)
- Anna Maria Di Giacomo
- University of Siena, Siena, Italy; Center for Immuno-Oncology. University Hospital of Siena, Viale Bracci, 16, Siena, Italy; NIBIT Foundation Onlus, Italy
| | - Michael Lahn
- IOnctura SA, Avenue Secheron 15, Geneva, Switzerland
| | - Alexander Mm Eggermont
- Princess Máxima Center and the University Medical Center Utrecht, Heidelberglaan 25, 3584 Utrecht, the Netherlands; Comprehensive Cancer Center Munich of the Technical University Munich and the Ludwig Maximiliaan University, Munich, Germany
| | - Bernard Fox
- Earle A. Chiles Research Institute at the Robert W. Franz Cancer Center, 4805 NE Glisan St. Suite 2N35 Portland, OR 97213, USA
| | - Ramy Ibrahim
- Parker Institute for Cancer Immunotherapy, 1 Letterman Drive, D3500, San Francisco, CA, USA
| | - Padmanee Sharma
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson, 1515 Holcombe Blvd, Houston, Texas 77030, USA
| | - James P Allison
- James P Allison Institute, MD Anderson, 1515 Holcombe Blvd, Texas 77030, USA
| | - Michele Maio
- University of Siena, Siena, Italy; Center for Immuno-Oncology. University Hospital of Siena, Viale Bracci, 16, Siena, Italy; NIBIT Foundation Onlus, Italy.
| |
Collapse
|
65
|
Tu C, Kulasinghe A, Barbour A, Souza-Fonseca-Guimaraes F. Leveraging spatial omics for the development of precision sarcoma treatments. Trends Pharmacol Sci 2024; 45:134-144. [PMID: 38212196 DOI: 10.1016/j.tips.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/13/2024]
Abstract
Sarcomas are rare and heterogeneous cancers that arise from bone or soft tissue, and are the second most prevalent solid cancer in children and adolescents. Owing to the complex nature of pediatric sarcomas, the development of therapeutics for pediatric sarcoma has seen little progress in the past decades. Existing treatments are largely limited to chemotherapy, radiation, and surgery. Limited knowledge of the sarcoma tumor microenvironment (TME) and of well-defined target antigens in the different subtypes necessitates an alternative investigative approach to improve treatments. Recent advances in spatial omics technologies have enabled a more comprehensive study of the TME in multiple cancers. In this opinion article we discuss advances in our understanding of the TME of some cancers enabled by spatial omics technologies, and we explore how these technologies might advance the development of precision treatments for sarcoma, especially pediatric sarcoma.
Collapse
Affiliation(s)
- Cui Tu
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Arutha Kulasinghe
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Andrew Barbour
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD 4102, Australia; Department of Surgery, Princess Alexandra Hospital, Brisbane, QLD 4102, Australia
| | | |
Collapse
|
66
|
Cao X, Ge Y, Yan Z, Hu X, Peng F, Zhang Y, He X, Zong D. MTDH enhances radiosensitivity of head and neck squamous cell carcinoma by promoting ferroptosis based on a prognostic signature. JOURNAL OF RADIATION RESEARCH 2024; 65:10-27. [PMID: 37981296 PMCID: PMC10803166 DOI: 10.1093/jrr/rrad074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/16/2023] [Indexed: 11/21/2023]
Abstract
Ionizing radiation (IR) induces ferroptosis in head and neck squamous cell carcinoma (HNSCC). But, it remains unclear whether ferroptosis affects the prognosis of HNSCC patients after receiving radiotherapy. This study aims to develop a ferroptosis signature to predict the radiosensitivity and prognosis of HNSCC. Ferroptosis-related genes, clinical data and RNA expression profiles were obtained from the FerrDb database, The Cancer Genome Atlas and GEO database. Prognostic genes were identified by random survival forest, univariate Cox regression, Kaplan-Meier and ROC analyses. Principal component analysis, multivariate Cox regression, nomogram and DCA analyses were conducted to estimate its predictive ability. Functional enrichment and immune-related analyses were performed to explore potential biological mechanisms and tumor immune microenvironment. The effect of the hub gene on ferroptosis and radiosensitivity was verified using flow cytometry, quantitative real-time PCR and clonogenic survival assay. We constructed a ferroptosis-related signature, including IL6, NCF2, metadherin (MTDH) and CBS. We classified patients into high-risk (HRisk) and low-risk groups according to the risk scores. The risk score was confirmed to be an independent predictor for overall survival (OS). Combining the clinical stage with the risk score, we established a predictive nomogram for OS. Furthermore, pathways related to tumorigenesis and tumor immune suppression were mainly enriched in HRisk. MTDH was verified to have a potent effect on IR-induced ferroptosis and consequently promoted radiosensitivity. We constructed a ferroptosis-related signature to predict radiosensitivity and OS in HNSCC patients. MTDH was identified as a promising therapeutic target in radioresistant HNSCC patients.
Collapse
Affiliation(s)
- Xiang Cao
- The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| | - Yizhi Ge
- The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| | - Zhenyu Yan
- The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| | - Xinyu Hu
- The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| | - Fanyu Peng
- The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| | - Yujie Zhang
- The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| | - Xia He
- The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu 210000, China
- Xuzhou Medical University, No. 209, Tongshan Road, Xuzhou, Jiangsu 221000, China
| | - Dan Zong
- The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| |
Collapse
|
67
|
Ou X, Zhai R, Wei W, Chen J, Ou D, Liao T, Xu T, Zhu Y, Wang Y, Huang S, Shi R, Wu B, Chen T, Li Y, Yang Z, Zhou C, Liu Y, Jiang Z, Zeng M, Liu X, Ji D, Ying H, Zhang Z, Hu C, Lu X, Ji Q, He X, Wang Y. Induction Toripalimab and Chemotherapy for Organ Preservation in Locally Advanced Laryngeal and Hypopharyngeal Cancer: A Single-Arm Phase II Clinical Trial. Clin Cancer Res 2024; 30:344-355. [PMID: 37955629 DOI: 10.1158/1078-0432.ccr-23-2398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/05/2023] [Accepted: 11/08/2023] [Indexed: 11/14/2023]
Abstract
PURPOSE The aim of this study was to assess the efficacy, toxicities, and potential role of larynx preservation of induction chemotherapy combined with programmed cell death protein 1 (PD-1) inhibitor in locally advanced laryngeal and hypopharyngeal cancer. PATIENTS AND METHODS This is a single-arm phase II study. Patients with histopathologically confirmed, resectable locally advanced laryngeal/hypopharyngeal squamous cell carcinoma and Eastern Cooperative Oncology Group Performance Status 0-1 were eligible. Three cycles of induction chemotherapy (paclitaxel 175 mg/m2 d1, cisplatin 25 mg/m2 d1-3) combined with PD-1 inhibitor (toripalimab 240 mg d0) were administered. Response assessment was performed after induction chemoimmunotherapy using RECIST 1.1 criteria. Patients with a complete/partial response of the primary tumor received concurrent chemoradiation, followed by maintenance therapy of toripalimab. Otherwise, patients were referred to surgery, followed by adjuvant (chemo) radiation and maintenance therapy of toripalimab. The primary endpoint is a larynx preservation rate at 3 months postradiation. RESULTS Twenty-seven patients were enrolled. Most cases exhibited stage IV disease (81.5%), with T4 representing 37.0%. Five patients underwent pretreatment tracheostomy because of impaired larynx function. Overall response rate of induction chemoimmunotherapy was 85.2%. At 3 months postradiation, the larynx preservation rate was 88.9%. With a median follow-up of 18.7 months, the 1-year overall survival rate, progression-free survival rate, and larynx preservation rate were 84.7%, 77.6%, and 88.7%, respectively. When excluding those with pretreatment tracheostomy, the 1-year larynx preservation rate was 95.5%. Exploratory analysis revealed that relapse correlated with enrichment of RNA signature of hypoxia and M2 macrophage-associated genes. CONCLUSIONS Induction toripalimab combined with chemotherapy provided encouraging activity, promising larynx preservation rate and acceptable toxicity in this cohort of extensively locally advanced laryngeal and hypopharyngeal cancer.
Collapse
Affiliation(s)
- Xiaomin Ou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ruiping Zhai
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wenjun Wei
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jiaying Chen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Dan Ou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Tian Liao
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Tingting Xu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yongxue Zhu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yulong Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shenglin Huang
- Shanghai Key Laboratory of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Rongliang Shi
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Bin Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Diagnostic Radiology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Tongzhen Chen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yuan Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhongyi Yang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Changming Zhou
- Department of Cancer Prevention, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yuan Liu
- Department of Endoscopy, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ziting Jiang
- Department of Endoscopy, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Min Zeng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Nursing Administration, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xin Liu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Head and Neck Tumors and Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Dongmei Ji
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Head and Neck Tumors and Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hongmei Ying
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Chaosu Hu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xueguan Lu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qinghai Ji
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiayun He
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yu Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
68
|
Muijlwijk T, Nijenhuis DNLM, Ganzevles SH, Brink A, Ke C, Fass JN, Rajamanickam V, Leemans CR, Koguchi Y, Fox BA, Poell JB, Brakenhoff RH, van de Ven R. Comparative analysis of immune infiltrates in head and neck cancers across anatomical sites. J Immunother Cancer 2024; 12:e007573. [PMID: 38212122 PMCID: PMC10806653 DOI: 10.1136/jitc-2023-007573] [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] [Accepted: 12/20/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND The response rate to immune checkpoint inhibitors targeting programmed cell death 1 (PD-1) receptor is 13%-18% for patients with recurrent or metastatic head and neck squamous cell carcinoma (HNSCC). Detailed understanding of the tumor immune microenvironment (TIME) is crucial in order to explain and improve this response rate. HNSCCs arise at various anatomical locations including the oral cavity, hypopharynx, larynx and oropharynx. Studies directly comparing immune infiltration between anatomical sites are scarce. Since the distinct locations could drive deviating microenvironments, we questioned whether the immune composition varies across these HNSCC sites. METHODS Here, we characterized the TIME of 76 fresh tumor specimens using flow cytometry and performed single-cell RNA-sequencing on nine head and neck tumor samples. RESULTS We found major differences in the composition of the TIME between patients. When comparing anatomical sites: tumors originating from the oral cavity had higher T cell infiltrates than tumors from other anatomical sites. The percentage of tumor-infiltrating T-lymphocytes positive for the immune checkpoint PD-1 varied considerably between patients, with the highest fraction of PD-1+ T cells found in larynx squamous cell carcinomas (SCCs). While we had hypothesized that the anatomical sites of tumor origin would drive sample clustering, our data showed that the type of TIME was more dominant and was particularly driven by the fraction of T cells positive for PD-1. Moreover, a high proportion of PD-1+ CD8+ T cells associated with an improved overall survival. Using single-cell RNA-sequencing, we observed that PD-1 expression was highest in the CD8-ENTPD1 tissue resident memory T cell/exhausted T cell and CD4-CXCL13 type 1 T helper cell clusters. CONCLUSIONS We found that oral cavity SCCs had the highest frequencies of T cells. We also observed considerable interpatient heterogeneity for PD-1 on T cells, with noticeably higher frequencies of PD-1+ CD4+ T helper cells in larynx SCCs. Within the entire cohort, a higher fraction of CD8+ T cells positive for PD-1 was linked to improved overall survival. Whether the fraction of PD-1+ T cells within the TIME enables immune checkpoint inhibitor response prediction for patients with head and neck cancer remains to be determined.
Collapse
Affiliation(s)
- Tara Muijlwijk
- Department of Otolaryngology/Head and Neck Surgery, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Dennis N L M Nijenhuis
- Department of Otolaryngology/Head and Neck Surgery, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Sonja H Ganzevles
- Department of Otolaryngology/Head and Neck Surgery, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Arjen Brink
- Department of Otolaryngology/Head and Neck Surgery, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Changlin Ke
- Department of Otolaryngology/Head and Neck Surgery, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Joseph N Fass
- Providence Cancer Institute, Earle A Chiles Research Institute, Portland, Oregon, USA
| | | | - C René Leemans
- Department of Otolaryngology/Head and Neck Surgery, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Yoshinobu Koguchi
- Providence Cancer Institute, Earle A Chiles Research Institute, Portland, Oregon, USA
| | - Bernard A Fox
- Providence Cancer Institute, Earle A Chiles Research Institute, Portland, Oregon, USA
| | - Jos B Poell
- Department of Otolaryngology/Head and Neck Surgery, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Ruud H Brakenhoff
- Department of Otolaryngology/Head and Neck Surgery, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Rieneke van de Ven
- Department of Otolaryngology/Head and Neck Surgery, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| |
Collapse
|
69
|
Feng Y, Guo K, Jin H, Jiang J, Wang M, Lin S. Adverse events of neoadjuvant combination immunotherapy for resectable cancer patients: a systematic review and meta-analysis. Front Immunol 2024; 14:1269067. [PMID: 38250059 PMCID: PMC10796654 DOI: 10.3389/fimmu.2023.1269067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 12/18/2023] [Indexed: 01/23/2024] Open
Abstract
Background Neoadjuvant combination immunotherapy is changing the treatment landscape for patients with cancer. Exploring the incidence of immune-related adverse events (irAEs) in relation to this novel approach may provide valuable insights for future clinical investigations. Methods This review was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. PubMed, Embase, Cochrane Library, American Society of Clinical Oncology (ASCO), and European Society of Medical Oncology (ESMO) websites were searched for all relevant literature from their inception to November 24, 2023. We then extracted the required data from the included studies and used the R software to analyze the pooled incidence of irAEs. Subgroup analyses examined the pooled incidence of irAEs according to cancer and combination types using a random-effects model. Results Sixteen studies involving 501 patients were included in the meta-analysis. Considering the heterogeneity of the study design, we analyzed the randomized controlled studies (RCTs) and the single-arm studies separately. In RCTs, the incidence of any-grade irAEs was 95.0% (95% confidence interval [CI] 87.3-99.3) and that of grade ≥3 irAEs was 24.0% (95% CI 13.7-36.0). In single-arm studies, the incidence of any-grade irAEs was 89.4% (95% CI 75.0-98.0) and grade ≥3 irAEs was 20.3% (95% CI 8.7-35.2). In both RCTs and single arms, the most common any- grade irAEs were rash and fatigue, while the most common grade ≥3 irAEs was abnormal liver function and colitis. Due to irAEs, 9.4% of patients in RCTs and 6.9% of patients in single-arm studies did not complete the prescribed neoadjuvant treatment cycle. Conclusion This study comprehensively summarized the incidence of irAEs in neoadjuvant combination immunotherapy. The occurrence of irAEs varies depending on the cancer and combination types. Our meta-analysis provides clinicians with essential guidance for the management of patients with cancer. Systematic review registration https://www.crd.york.ac.uk/prospero, identifier CRD42023387969.
Collapse
Affiliation(s)
- Yuqian Feng
- Hangzhou School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Kaibo Guo
- Department of Oncology, Hangzhou First People’s Hospital, Hangzhou, Zhejiang, China
| | - Huimin Jin
- Department of Oncology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jing Jiang
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Menglei Wang
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shengyou Lin
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
70
|
Deutsch JS, Cimino-Mathews A, Thompson E, Provencio M, Forde PM, Spicer J, Girard N, Wang D, Anders RA, Gabrielson E, Illei P, Jedrych J, Danilova L, Sunshine J, Kerr KM, Tran M, Bushong J, Cai J, Devas V, Neely J, Balli D, Cottrell TR, Baras AS, Taube JM. Association between pathologic response and survival after neoadjuvant therapy in lung cancer. Nat Med 2024; 30:218-228. [PMID: 37903504 PMCID: PMC10803255 DOI: 10.1038/s41591-023-02660-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 10/23/2023] [Indexed: 11/01/2023]
Abstract
Neoadjuvant immunotherapy plus chemotherapy improves event-free survival (EFS) and pathologic complete response (0% residual viable tumor (RVT) in primary tumor (PT) and lymph nodes (LNs)), and is approved for treatment of resectable lung cancer. Pathologic response assessment after neoadjuvant therapy is the potential analog to radiographic response for advanced disease. However, %RVT thresholds beyond pathologic complete response and major pathologic response (≤10% RVT) have not been explored. Pathologic response was prospectively assessed in the randomized, phase 3 CheckMate 816 trial (NCT02998528), which evaluated neoadjuvant nivolumab (anti-programmed death protein 1) plus chemotherapy in patients with resectable lung cancer. RVT, regression and necrosis were quantified (0-100%) in PT and LNs using a pan-tumor scoring system and tested for association with EFS in a prespecified exploratory analysis. Regardless of LN involvement, EFS improved with 0% versus >0% RVT-PT (hazard ratio = 0.18). RVT-PT predicted EFS for nivolumab plus chemotherapy (area under the curve = 0.74); 2-year EFS rates were 90%, 60%, 57% and 39% for patients with 0-5%, >5-30%, >30-80% and >80% RVT, respectively. Each 1% RVT associated with a 0.017 hazard ratio increase for EFS. Combining pathologic response from PT and LNs helped differentiate outcomes. When compared with radiographic response and circulating tumor DNA clearance, %RVT best approximated EFS. These findings support pathologic response as an emerging survival surrogate. Further assessment of the full spectrum of %RVT in lung cancer and other tumor types is warranted. ClinicalTrials.gov registration: NCT02998528 .
Collapse
Affiliation(s)
- Julie Stein Deutsch
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ashley Cimino-Mathews
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth Thompson
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Patrick M Forde
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Nicolas Girard
- Institut du Thorax Curie-Montsouris, Institut Curie, Paris, France
| | - Daphne Wang
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert A Anders
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Edward Gabrielson
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter Illei
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jaroslaw Jedrych
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ludmila Danilova
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joel Sunshine
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Mia Tran
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | | | | | | | - Alex S Baras
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Janis M Taube
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
71
|
Cai F, Xu H, Song S, Wang G, Zhang Y, Qian J, Xu L. Knockdown of Ubiquitin-Conjugating Enzyme E2 T Abolishes the Progression of Head and Neck Squamous Cell Carcinoma by Inhibiting NF-Κb Signaling and inducing Ferroptosis. Curr Protein Pept Sci 2024; 25:577-585. [PMID: 38584528 DOI: 10.2174/0113892037287640240322084946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/17/2024] [Accepted: 02/23/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Ubiquitin-conjugating enzyme 2T (UBE2T) has been reported to be associated with uncontrolled cell growth and tumorigenesis in multiple cancer types. However, the understanding of its regulatory role in the carcinogenesis of Head And Neck Squamous Cell Carcinoma (HNSC) is limited. METHODS UBE2T expression in HNSC patient samples and the correlation between its expression and patients' survival rates were evaluated using The Cancer Genome Atlas (TCGA) database. Cell survival and proliferation were investigated in UM-SCC1 and UM-SCC15 cells infected with control and shUBE2T lentivirus. The xenograft mouse model was established using UM-SCC15 cells to examine HNSC tumorigenesis with or without UBE2T. Western blot, qRT-PCR, and ferroptosis assays were carried out to disclose the interaction between UBE2T and NF-κB signaling and ferroptosis. RESULTS The increased expression of UBE2T was noted in tumor tissues of patients with HNSC, correlating with a significantly reduced overall survival time in this patient cohort. Knockdown of UBE2T inhibited HNSC tumorigenesis and tumor growth. Mechanistically, inhibition of UBE2T suppressed NF-κB signaling and induced ferroptosis in HNSC. CONCLUSION Our study underscores the multifaceted role of UBE2T in HNSC, illuminating its potential as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Feng Cai
- Department of Radiation Oncology, the First Affiliated Hospital of Bengbu Medical College, Zhihuai Road, Bengbu, 233000, Anhui, China
| | - Hongbo Xu
- Department of Radiation Oncology, the First Affiliated Hospital of Bengbu Medical College, Zhihuai Road, Bengbu, 233000, Anhui, China
| | - Shilong Song
- Department of Radiation Oncology, the First Affiliated Hospital of Bengbu Medical College, Zhihuai Road, Bengbu, 233000, Anhui, China
| | - Gengming Wang
- Department of Radiation Oncology, the First Affiliated Hospital of Bengbu Medical College, Zhihuai Road, Bengbu, 233000, Anhui, China
| | - Yajun Zhang
- Department of Radiation Oncology, the First Affiliated Hospital of Bengbu Medical College, Zhihuai Road, Bengbu, 233000, Anhui, China
| | - Jing Qian
- Department of Radiation Oncology, the First Affiliated Hospital of Bengbu Medical College, Zhihuai Road, Bengbu, 233000, Anhui, China
| | - Lu Xu
- Department of Radiation Oncology, the First Affiliated Hospital of Bengbu Medical College, Zhihuai Road, Bengbu, 233000, Anhui, China
| |
Collapse
|
72
|
Navran A, Al-Mamgani A, Elzinga H, Kessels R, Vens C, Tesselaar M, van den Brekel M, de Haan R, van Triest B, Verheij M. Phase I feasibility study of Olaparib in combination with loco-regional radiotherapy in head and neck squamous cell carcinoma. Clin Transl Radiat Oncol 2024; 44:100698. [PMID: 38021094 PMCID: PMC10654000 DOI: 10.1016/j.ctro.2023.100698] [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: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose PARP-inhibitors have potent radiosensitizing properties in pre-clinical models. To identify the maximum tolerated dose (MTD) of the PARP-inhibitor Olaparib in combination with radiotherapy in patients with head and neck cancer, a single institutional phase-I dose escalation trial was initiated. Patients and methods The starting dose of Olaparib was 25 mg BID, combined with radiotherapy (70 Gy in 35 fractions). The MTD was defined as the highest dose-level at which not more than 20 % of patients experience dose-limiting toxicities (DLT) or as the highest reached dose in the absence of DLT's. Results One week Olaparib-only treatment (25 mg QD) was administered to all patients prior to the start of radiotherapy. In dose-level I, Olaparib (25 mg BID) was combined with accelerated radiotherapy (70 Gy in 6 weeks). Because of DLT's in 3 of the 4 treated patients (acute tracheotomy 5 and 7 months and osteoradionecrosis 7 months after treatment), the Olaparib dose was de-escalated to 25 mg QD, and combined with conventional radiotherapy (70 Gy in 7 weeks) (dose-level II). There were no DLT's observed in 5 patients treated within dose-level II. After a median follow-up of 60 months, the 4-year LRC and OS rates were 77.8 % and 88.9 %, respectively. Conclusion Olaparib 25 mg QD combined with conventionally fractionated radiotherapy was well tolerated and identified as the MTD while severe DLT's were observed when Olaparib 25 mg BID was combined with accelerated radiation. This combination might be further explored in future Olaparib dose escalation studies in patients with locally-advanced HNSCC unfit for cisplatin-based chemoradiotherapy.
Collapse
Affiliation(s)
- Arash Navran
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Abrahim Al-Mamgani
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hester Elzinga
- Department of Head and Neck Surgery, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rob Kessels
- Department of Biomerics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Conchita Vens
- Department of Head and Neck Surgery, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Margot Tesselaar
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Michiel van den Brekel
- Department of Head and Neck Surgery, Netherlands Cancer Institute and Department of Oral and Maxillo-Facial Surgery, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Rosemarie de Haan
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Baukelien van Triest
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marcel Verheij
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
73
|
Trinh JQ, Acosta C, Easwar A, Galamaga R, Tan A. Durable and dramatic response to checkpoint inhibition combined with COX-2 inhibitor celecoxib in a patient with p16+ metastatic sinonasal undifferentiated carcinoma: A case study. Cancer Rep (Hoboken) 2024; 7:e1915. [PMID: 37867289 PMCID: PMC10809189 DOI: 10.1002/cnr2.1915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 10/01/2023] [Accepted: 10/08/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND Sinonasal undifferentiated carcinoma (SNUC) is an exceedingly rare head and neck malignancy. No consensus exists on treatment for metastatic disease. CASE A 56-year-old female was diagnosed with SNUC after endorsing sinus congestion, diplopia, and right orbital pain. Initially treated with surgery and radiation, she later developed significant metastatic disease. She demonstrated progression of her hepatic metastases under pembrolizumab therapy. However, the addition of ipilimumab and a COX-2 inhibitor resulted in significant improvement in her lesions as well as an ongoing durable response. Her regimen was complicated by immune-related adverse events successfully treated with steroids. CONCLUSION Dual checkpoint inhibition deserves consideration when treating metastatic SNUC, especially after single agent therapy has failed. The positive effect of this treatment may be augmented by IDO1 inhibition.
Collapse
Affiliation(s)
- Jonathan Q. Trinh
- Department of Internal MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Cassaundra Acosta
- Department of Medical Oncology and HematologyCity of HopeGoodyearArizonaUSA
| | - Arti Easwar
- Department of Pathology and Laboratory MedicineCity of HopeGoodyearArizonaUSA
| | - Robert Galamaga
- Department of Medical Oncology and HematologyCity of HopeGoodyearArizonaUSA
| | - Alan Tan
- Department of Internal MedicineRush University Medical CenterChicagoIllinoisUSA
| |
Collapse
|
74
|
Stockem CF, Galsky MD, van der Heijden MS. Turning up the heat: CTLA4 blockade in urothelial cancer. Nat Rev Urol 2024; 21:22-34. [PMID: 37608154 DOI: 10.1038/s41585-023-00801-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2023] [Indexed: 08/24/2023]
Abstract
Anti-PD1 and anti-PDL1 monotherapies have shown clinical efficacy in stage IV urothelial cancer and are integrated into current clinical practice. However, only a small number of the patients treated with single-agent checkpoint blockade experience an antitumour response. Insufficient priming or inhibitory factors in the tumour immune microenvironment might have a role in the lack of response. CTLA4 is an inhibitory checkpoint on activated T cells that is being studied as a therapeutic target in combination with anti-PD1 or anti-PDL1 therapies in advanced urothelial cancer. In locally advanced urothelial cancer, this combination approach has shown encouraging antitumour effects when administered pre-operatively. We believe that the presence of pre-existing intratumoural T cell immunity is not a prerequisite for response to combination therapy and that the additional value of CTLA4 blockade might involve the broadening of peripheral T cell priming, thereby transforming immunologically cold tumours into hot tumours.
Collapse
Affiliation(s)
- Chantal F Stockem
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Matthew D Galsky
- Department of Genitourinary Medical Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | | |
Collapse
|
75
|
Smussi D, Mattavelli D, Paderno A, Gurizzan C, Lorini L, Romani C, Bignotti E, Grammatica A, Ravanelli M, Bossi P. Revisiting the concept of neoadjuvant and induction therapy in head and neck cancer with the advent of immunotherapy. Cancer Treat Rev 2023; 121:102644. [PMID: 37862833 DOI: 10.1016/j.ctrv.2023.102644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 10/03/2023] [Accepted: 10/11/2023] [Indexed: 10/22/2023]
Abstract
The treatment of locally advanced (LA) Head and Neck Squamous Cell Carcinoma (HNSCC) is based on surgery followed by (chemo)radiation or on curative (chemo)radiation, depending on site and stage. Despite optimal locoregional treatment, about 50% of patients recur, with a huge impact on prognosis and substantial morbidity. The advent of immunotherapy (IT) with immune checkpoint inhibitors (ICIs) changed the paradigm of systemic treatment for recurrent/metastatic (RM) disease, showing activity, efficacy, and safety in both platinum-resistant and platinum-naïve patients. Such data led clinicians to design clinical trials to investigate early administration of IT even in the neoadjuvant or window of opportunity setting. In this review, we examine the published and ongoing trials investigating IT in the neoadjuvant setting for LA HNSCC. We address the current challenges of this treatment modality: optimal patient selection for neoadjuvant IT; choosing the appropriate systemic approach to enhance response without compromising tolerability; determining the ideal study endpoint, with a focus on major pathological response as a potential surrogate for overall survival; evaluating treatment response through imaging, considering the discordance between radiological and pathological assessments; and the influence of neoadjuvant IT response on locoregional treatment de-escalation strategies.
Collapse
Affiliation(s)
- Davide Smussi
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, ASST-Spedali Civili, Brescia, Italy
| | - Davide Mattavelli
- Otorhinolaryngology - Head and Neck Surgery Unit, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, ASST-Spedali Civili, Brescia, Italy
| | - Alberto Paderno
- Otorhinolaryngology Unit, IRCCS Humanitas Research Hospital, Rozzano, MI, Italy
| | - Cristina Gurizzan
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, ASST-Spedali Civili, Brescia, Italy
| | - Luigi Lorini
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, MI, Italy
| | - Chiara Romani
- Angelo Nocivelli Institute of Molecular Medicine, University of Brescia and ASST Spedali Civili di Brescia, Brescia, Italy
| | - Eliana Bignotti
- Angelo Nocivelli Institute of Molecular Medicine, University of Brescia and ASST Spedali Civili di Brescia, Brescia, Italy
| | - Alberto Grammatica
- Otorhinolaryngology - Head and Neck Surgery Unit, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, ASST-Spedali Civili, Brescia, Italy
| | - Marco Ravanelli
- Radiology Unit, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health University of Brescia, ASST-Spedali Civili, Brescia, Italy
| | - Paolo Bossi
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, MI, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy.
| |
Collapse
|
76
|
Xu P, Fang Q, Zhao Z, Cao F, Wu D, Liu X. Evaluation of neoadjuvant chemotherapy combined with PD-1 inhibitors in patients with oropharyngeal and hypopharyngeal squamous cell carcinoma: a comparative study of antitumor activity. Cancer Immunol Immunother 2023; 72:4209-4219. [PMID: 37837458 PMCID: PMC10991092 DOI: 10.1007/s00262-023-03557-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 09/28/2023] [Indexed: 10/16/2023]
Abstract
PURPOSE To assess the antitumor activity of neoadjuvant chemotherapy in conjunction with PD-1 inhibitors (neoadjuvant chemoimmunotherapy) among patients with oropharyngeal and hypopharyngeal squamous cell carcinoma (OPHSCC) and compare its efficacy with neoadjuvant chemotherapy alone. METHODS We conducted a retrospective analysis using data from patients diagnosed with OPHSCC and treated at the Sun Yat-sen University Cancer Center between September 2012 and August 2022. We included patients who received neoadjuvant chemotherapy alone or combined with PD-1 inhibitors. We assessed the clinical response using the Response Evaluation Criteria in Solid Tumors and evaluated progression-free survival (PFS) and overall survival (OS). RESULTS Preliminary results demonstrate that neoadjuvant chemoimmunotherapy exhibited robust antitumor activity in OPHSCC, with an impressive overall response rate (ORR) of 81.0%. Complete response and partial response rates were 14.9% and 65.9%, respectively. Notably, neoadjuvant chemoimmunotherapy demonstrated superior PFS and OS to neoadjuvant chemotherapy alone. The 1-year PFS rate was 80.7%, and the 2-year rate was 61.1%. Additionally, the 1-year OS rate reached 92.3%. Finally, a multivariate analysis identified the American Joint Committee on Cancer stage reduction post-treatment as a favorable predictor of PFS. CONCLUSION Our results underscore the promising potential of neoadjuvant chemoimmunotherapy in enhancing antitumor activity in patients with OPHSCC. The robust ORR, along with improved PFS and OS, supports the utility of this combined approach. These results pave the way for further investigations to validate and refine the application of neoadjuvant chemoimmunotherapy in this challenging clinical context.
Collapse
Affiliation(s)
- Pengfei Xu
- Department of Head and Neck Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
- State Key Laboratory of Oncology in South China, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
- Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Qi Fang
- Department of Head and Neck Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
- State Key Laboratory of Oncology in South China, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
- Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Zheng Zhao
- Department of Head and Neck Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
- State Key Laboratory of Oncology in South China, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
- Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Fei Cao
- Department of Head and Neck Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
- State Key Laboratory of Oncology in South China, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
- Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Di Wu
- Department of Head and Neck Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China.
- State Key Laboratory of Oncology in South China, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China.
- Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China.
| | - Xuekui Liu
- Department of Head and Neck Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China.
- State Key Laboratory of Oncology in South China, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China.
- Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China.
| |
Collapse
|
77
|
Reticker-Flynn NE, Engleman EG. Lymph nodes: at the intersection of cancer treatment and progression. Trends Cell Biol 2023; 33:1021-1034. [PMID: 37149414 PMCID: PMC10624650 DOI: 10.1016/j.tcb.2023.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 05/08/2023]
Abstract
Metastasis to lymph nodes (LNs) is a common feature of disease progression in most solid organ malignancies. Consequently, LN biopsy and lymphadenectomy are common clinical practices, not only because of their diagnostic utility but also as a means of deterring further metastatic spread. LN metastases have the potential to seed additional tissues and can induce metastatic tolerance, a process by which tumor-specific immune tolerance in LNs promotes further disease progression. Nonetheless, phylogenetic studies have revealed that distant metastases are not necessarily derived from nodal metastases. Furthermore, immunotherapy efficacy is increasingly being attributed to initiation of systemic immune responses within LNs. We argue that lymphadenectomy and nodal irradiation should be approached with caution, particularly in patients receiving immunotherapy.
Collapse
Affiliation(s)
- Nathan E Reticker-Flynn
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Edgar G Engleman
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
78
|
Wu WJ, Liu Q, An PG, Wang L, Zhang JY, Chen Y, Zhang T, Zhang J. Neoadjuvant tislelizumab combined with chemotherapy in locally advanced oral or oropharyngeal squamous cell carcinoma: a real-world retrospective study. Front Immunol 2023; 14:1282629. [PMID: 38035079 PMCID: PMC10685444 DOI: 10.3389/fimmu.2023.1282629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Objectives The treatment of locally advanced oral or oropharyngeal squamous cell carcinoma (LAOOPSCC) is surgery and radiotherapy or chemoradiotherapy but with unsatisfactory survival rate. Neoadjuvant programmed death-1 (PD-1) therapy are being used in several clinical trials. Therefore, in this retrospective study we aimed to determine the feasibility of neoadjuvant tislelizumab plus chemotherapy followed by surgery for LAOOPSCC. Materials and methods The clinical data of 33 patients with LAOOPSCC who received neoadjuvant PD-1 inhibitors and chemotherapy between April 2021 and October 2022 were retrospectively analyzed. Patients with stage III-IV LAOOPSCC received tislelizumab, albumin-bound paclitaxel, and cisplatin every 3 weeks (Q3W) for two cycles, followed by surgery and adjuvant radiotherapy or concurrent chemoradiotherapy. A median follow-up period was 20 months. Results The objective response rate (ORR) was 66.7%, with the major pathological response (MPR) rate at 54.5%, and the pathological complete response (pCR) rate was 33.3%. Sixteen patients underwent limited surgeries, and 15 patients were remitted from undergoing mandibulectomy and 9 patients were remitted from undergoing near total glossectomy or total glossectomy. A significant difference in the overall survival (OS) and disease-free survival (DFS) was observed in patients who achieved major pathological response (MPR) than who did not. The most common adverse events in neoadjuvant therapy were alopecia, decreased appetite or anorexia, leukopenia, and fatigue. Conclusion Neoadjuvant PD-1 inhibitors combined with chemotherapy are feasible and safe, with a high pathological response and possible organ preservation in oral or oropharyngeal squamous cell carcinoma. However, further studies with a larger cohort of patients and longer follow-up period is required to strengthen our findings and evaluate the survival benefits of the treatment.
Collapse
Affiliation(s)
- Wen-Jie Wu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- National Center of Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Qian Liu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- National Center of Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Pu-Gen An
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- National Center of Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Lin Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- National Center of Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Jian-Yun Zhang
- National Center of Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yan Chen
- National Center of Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Tong Zhang
- Departmet of Oncology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- National Center of Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
79
|
Hoeijmakers LL, Reijers ILM, Blank CU. Biomarker-Driven Personalization of Neoadjuvant Immunotherapy in Melanoma. Cancer Discov 2023; 13:2319-2338. [PMID: 37668337 DOI: 10.1158/2159-8290.cd-23-0352] [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: 03/24/2023] [Revised: 06/27/2023] [Accepted: 07/26/2023] [Indexed: 09/06/2023]
Abstract
The introduction of immunotherapy has ushered in a new era of anticancer therapy for many cancer types including melanoma. Given the increasing development of novel compounds and combinations and the investigation in earlier disease stages, the need grows for biomarker-based treatment personalization. Stage III melanoma is one of the front-runners in the neoadjuvant immunotherapy field, facilitating quick biomarker identification by its immunogenic capacity, homogeneous patient population, and reliable efficacy readout. In this review, we discuss potential biomarkers for response prediction to neoadjuvant immunotherapy, and how the neoadjuvant melanoma platform could pave the way for biomarker identification in other tumor types. SIGNIFICANCE In accordance with the increasing rate of therapy development, the need for biomarker-driven personalized treatments grows. The current landscape of neoadjuvant treatment and biomarker development in stage III melanoma can function as a poster child for these personalized treatments in other tumors, assisting in the development of new biomarker-based neoadjuvant trials. This will contribute to personalized benefit-risk predictions to identify the most beneficial treatment for each patient.
Collapse
Affiliation(s)
- Lotte L Hoeijmakers
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam, the Netherlands
| | - Irene L M Reijers
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam, the Netherlands
| | - Christian U Blank
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam, the Netherlands
- Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
- Molecular Oncology and Immunology, Netherlands Cancer Institute (NKI), Amsterdam, the Netherlands
| |
Collapse
|
80
|
Huang S, Xiong C, Tan K. Neoadjuvant PD-1/PD-L1 axis blockade for patients with head and neck squamous cell carcinoma. Am J Otolaryngol 2023; 44:103985. [PMID: 37442083 DOI: 10.1016/j.amjoto.2023.103985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/04/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a common type of cancer, and approximately 64 % are in a locally advanced stage at diagnosis. Therefore, neoadjuvant therapy is of great importance. However, traditional neoadjuvant strategies for HNSCC have shown limited efficacy and high complications. And it is urgent to explore new neoadjuvant approaches. With the breakthrough progress of PD-1/PD-L1 axis blockade in recurrent/metastatic HNSCC, neoadjuvant PD-1/PD-L1 axis blockade is gradually showing positive prospects for HNSCC. This study found that the combination of PD-1/PD-L1 axis blockade and chemotherapy or radiotherapy are potential with the overall response rate (ORR) of 45.0 %-96.7 % and 47.6 %-56.7 %, the pathological complete response (pCR) of 16.7 %-42.3 % and 33.3 %-100.0 %, and the main pathological response (MPR) of 26.9 %-74.1 % and 60.0 %-100.0 %, respectively. But the combination of PD-1/PD-L1 axis blockade and CTLA-4 blockade is worth questioning. And we also found pCR and MPR can be early indicators for long-term prognosis and provide five directions for neoadjuvant PD-1/PD-L1 axis blockade in the future.
Collapse
Affiliation(s)
- Shuang Huang
- Department of Stomatology, The Affiliated Shapingba Hospital of Chongqing University, Chongqing, China.
| | - Chuang Xiong
- Department of Bone and Soft Tissue Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Kui Tan
- Department of Stomatology, Chongqing University Jiangjin Hospital, Chongqing, China
| |
Collapse
|
81
|
Fridman WH, Meylan M, Pupier G, Calvez A, Hernandez I, Sautès-Fridman C. Tertiary lymphoid structures and B cells: An intratumoral immunity cycle. Immunity 2023; 56:2254-2269. [PMID: 37699391 DOI: 10.1016/j.immuni.2023.08.009] [Citation(s) in RCA: 73] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 09/14/2023]
Abstract
The generation of anti-tumor immunity in the draining lymph nodes is known as the cancer immunity cycle. Accumulating evidence supports the occurrence of such a cycle at tumor sites in the context of chronic inflammation. Here, we review the role of tertiary lymphoid structures (TLS) in the generation of T and B cell immunities, focusing on the impact of B cells that undergo full maturation, resulting in the generation of plasma cells (PCs) producing high-affinity IgG and IgA antibodies. In this context, we propose that antibodies binding to tumor cells induce macrophage or natural killer (NK)-cell-dependent apoptosis. Subsequently, released antigen-antibody complexes are internalized and processed by dendritic cells (DCs), amplifying antigen presentation to T cells. Immune complexes may also be fixed by follicular DCs (FDCs) in TLS, thereby increasing memory B cell responses. This amplification loop creates an intra-tumoral immunity cycle, capable of increasing sensitivity of tumors to immunotherapy even in cancers with low mutational burden.
Collapse
Affiliation(s)
- Wolf H Fridman
- Centre de Recherche des Cordeliers, INSERM U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France; Equipe labellisée Ligue Contre le Cancer (EL 2021), Paris, France.
| | - Maxime Meylan
- Centre de Recherche des Cordeliers, INSERM U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France; Equipe labellisée Ligue Contre le Cancer (EL 2021), Paris, France
| | - Guilhem Pupier
- Centre de Recherche des Cordeliers, INSERM U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France; Equipe labellisée Ligue Contre le Cancer (EL 2021), Paris, France
| | - Anne Calvez
- Centre de Recherche des Cordeliers, INSERM U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France; Equipe labellisée Ligue Contre le Cancer (EL 2021), Paris, France
| | - Isaïas Hernandez
- Centre de Recherche des Cordeliers, INSERM U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France; Equipe labellisée Ligue Contre le Cancer (EL 2021), Paris, France
| | - Catherine Sautès-Fridman
- Centre de Recherche des Cordeliers, INSERM U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France; Equipe labellisée Ligue Contre le Cancer (EL 2021), Paris, France
| |
Collapse
|
82
|
van der Leun AM, Traets JJ, Vos JL, Elbers JB, Patiwael S, Qiao X, Machuca-Ostos M, Thommen DS, Haanen JB, Schumacher TN, Zuur CL. Dual Immune Checkpoint Blockade Induces Analogous Alterations in the Dysfunctional CD8+ T-cell and Activated Treg Compartment. Cancer Discov 2023; 13:2212-2227. [PMID: 37548431 PMCID: PMC10551666 DOI: 10.1158/2159-8290.cd-22-0851] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 05/02/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
To dissect the effect of neoadjuvant PD-1 and CTLA4 blockade on intratumoral T cells in treatment-naive head and neck squamous cell carcinoma, we analyzed primary tumor immune infiltrates from responding and nonresponding patients. At baseline, a higher ratio between active (4-1BB/OX40+) and inactive regulatory CD4+ T cells was associated with immunotherapy response. Furthermore, upon therapy, this active regulatory T-cell (Treg) population showed a profound decrease in responding patients. In an analogous process, intratumoral dysfunctional CD8+ T cells displayed decreased expression of activity and dysfunction-related genes in responding patients, whereas in clinical nonresponders, natural killer cells showed an increased cytotoxic profile early upon treatment. These data reveal immunologic changes in response to dual PD-1/CTLA4 blockade, including a parallel remodeling of presumed tumor-reactive Treg and CD8+ T-cell compartments in responding patients, and indicate that the presence of activated Tregs at baseline may be associated with response. SIGNIFICANCE In head and neck squamous cell carcinoma, neoadjuvant PD-1/CTLA4 blockade has shown substantial response rates (20%-35%). As recognition of tumor antigens by T cells appears to be a critical driver of therapy response, a better understanding of alterations in T-cell state that are associated with response and resistance is of importance. This article is featured in Selected Articles from This Issue, p. 2109.
Collapse
Affiliation(s)
- Anne M. van der Leun
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Joleen J.H. Traets
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Joris L. Vos
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Joris B.W. Elbers
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sanne Patiwael
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Xiaohang Qiao
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Mercedes Machuca-Ostos
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Daniela S. Thommen
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - John B.A.G. Haanen
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ton N.M. Schumacher
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - Charlotte L. Zuur
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Otorhinolaryngology Head and Neck Surgery, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
83
|
Valero C, Golkaram M, Vos JL, Xu B, Fitzgerald C, Lee M, Kaplan S, Han CY, Pei X, Sarkar R, Boe LA, Pandey A, Koh ES, Zuur CL, Solit DB, Pawlowski T, Liu L, Ho AL, Chowell D, Riaz N, Chan TA, Morris LG. Clinical-genomic determinants of immune checkpoint blockade response in head and neck squamous cell carcinoma. J Clin Invest 2023; 133:e169823. [PMID: 37561583 PMCID: PMC10541199 DOI: 10.1172/jci169823] [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: 02/16/2023] [Accepted: 08/03/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUNDRecurrent and/or metastatic (R/M) head and neck squamous cell carcinoma (HNSCC) is generally an incurable disease, with patients experiencing median survival of under 10 months and significant morbidity. While immune checkpoint blockade (ICB) drugs are effective in approximately 20% of patients, the remaining experience limited clinical benefit and are exposed to potential adverse effects and financial costs. Clinically approved biomarkers, such as tumor mutational burden (TMB), have a modest predictive value in HNSCC.METHODSWe analyzed clinical and genomic features, generated using whole-exome sequencing, in 133 ICB-treated patients with R/M HNSCC, of whom 69 had virus-associated and 64 had non-virus-associated tumors.RESULTSHierarchical clustering of genomic data revealed 6 molecular subtypes characterized by a wide range of objective response rates and survival after ICB therapy. The prognostic importance of these 6 subtypes was validated in an external cohort. A random forest-based predictive model, using several clinical and genomic features, predicted progression-free survival (PFS), overall survival (OS), and response with greater accuracy than did a model based on TMB alone. Recursive partitioning analysis identified 3 features (systemic inflammatory response index, TMB, and smoking signature) that classified patients into risk groups with accurate discrimination of PFS and OS.CONCLUSIONThese findings shed light on the immunogenomic characteristics of HNSCC tumors that drive differential responses to ICB and identify a clinical-genomic classifier that outperformed the current clinically approved biomarker of TMB. This validated predictive tool may help with clinical risk stratification in patients with R/M HNSCC for whom ICB is being considered.FUNDINGFundación Alfonso Martín Escudero, NIH R01 DE027738, US Department of Defense CA210784, The Geoffrey Beene Cancer Research Center, The MSKCC Population Science Research Program, the Jayme Flowers Fund, the Sebastian Nativo Fund, and the NIH/NCI Cancer Center Support Grant P30 CA008748.
Collapse
Affiliation(s)
- Cristina Valero
- Head and Neck Service, Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | | | - Joris L. Vos
- Head and Neck Service, Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Bin Xu
- Department of Pathology and Laboratory Medicine
| | - Conall Fitzgerald
- Head and Neck Service, Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Mark Lee
- Head and Neck Service, Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | | | - Catherine Y. Han
- Head and Neck Service, Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Xin Pei
- Department of Radiation Oncology, and
| | | | - Lillian A. Boe
- Department of Biostatistics and Epidemiology, MSKCC, New York, New York, USA
| | - Abhinav Pandey
- Head and Neck Service, Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Elizabeth S. Koh
- Head and Neck Service, Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Charlotte L. Zuur
- Department of Head and Neck Oncology and Surgery, Antoni van Leeuwenhoek Hospital–Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Otorhinolaryngology and Head and Neck Surgery, Leiden University Medical Center, Leiden, Netherlands
| | | | | | - Li Liu
- Illumina Inc., San Diego, California, USA
| | - Alan L. Ho
- Department of Medicine, MSKCC, New York, New York, USA
| | - Diego Chowell
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Timothy A. Chan
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Luc G.T. Morris
- Head and Neck Service, Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| |
Collapse
|
84
|
Chen S, Yang Y, Wang R, Fang J. Neoadjuvant PD-1/PD-L1 inhibitors combined with chemotherapy had a higher ORR than mono-immunotherapy in untreated HNSCC: Meta-analysis. Oral Oncol 2023; 145:106479. [PMID: 37478574 DOI: 10.1016/j.oraloncology.2023.106479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND HNSCC is one of the most common types of cancer worldwide and immune checkpoint inhibitor has shown favorable therapeutic effect in R/M HNSC. However, the application of immunotherapy in untreated HNSCC still needs to be discovered since most R/M HNSCC patients have been treated before and their drug susceptibility and immune tumor microenvironment have changed. This meta-analysis tries to compare immunotherapy and immunochemotherapy in untreated HNSCC and give a reference for clinic application. METHODS Electronic databases, including PubMed, Embase, and Web of Science, were systematically searched from inception through August 31, 2022. The primary outcomes were efficacy, evaluated by objective response rate, 1-year OS and 1-year PFS, and safety, evaluated by grade 3-4 adverse reaction rate. RESULTS A total of 1092 patients from twenty-four studies were included, 282 (25.8%) of which had ORR reported. The average ORR was 37% (95%CI = 26%-49%). Immunochemotherapy could have higher ORR than immunotherapy patients (ORR: 61% vs 22%), and favorable 1-year overall survival from PD-L1 inhibitor (OS = 84%, 95%CI 76%-93%). Radiotherapy after neoadjuvant immunotherapy was equal with the other treatments like chemotherapy and surgery (84% vs 88%, subgroup df p = 0.7). There was no apparent difference between immunotherapy and immunochemotherapy (32% vs 42%, subgroup df p = 0.60). CONCLUSION HNSCC patients could benefit more from neoadjuvant immunochemotherapy.
Collapse
Affiliation(s)
- Shaoshi Chen
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yifan Yang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ru Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jugao Fang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
85
|
Ren S, Lan T, Wu F, Chen S, Jiang X, Huo C, Li Z, Xie S, Wu D, Wang R, Li Y, Qiu L, Huang G, Li S, Wang X, Cen M, Cai T, Lin Z, Li J, Li B. Intratumoral CD103 + CD8 + T cells predict response to neoadjuvant chemoimmunotherapy in advanced head and neck squamous cell carcinoma. Cancer Commun (Lond) 2023; 43:1143-1163. [PMID: 37658605 PMCID: PMC10565384 DOI: 10.1002/cac2.12480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/19/2023] [Accepted: 08/22/2023] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND Immune cell heterogenicity is known to determine the therapeutic response to cancer progression. Neoadjuvant chemoimmunotherapy (NACI) has shown clinical benefits in some patients with advanced head and neck squamous cell carcinoma (HNSCC), but the underlying mechanism behind this clinical response is unknown. The efficacy of NACI needs to be potentiated by identifying accurate biomarkers to predict clinical responses. Here, we attempted to identify molecules predicting NACI response in advanced HNSCC. METHODS We performed combined single-cell RNA sequencing (scRNA-seq) and multiplex immunofluorescence (mIHC) staining with tumor samples derived from NACI-treated HNSCC patients to identify a new tumor-infiltrating cell (TIL) subtype, CD103+ CD8+ TILs, associated with clinical response, while both in vitro and in vivo assays were carried out to determine its antitumor efficiency. The regulatory mechanism of the CD103+ CD8+ TILs population was examined by performing cell-cell interaction analysis of the scRNA-seq data and spatial analysis of the mIHC images. RESULTS We established intratumoral CD103+ CD8+ TILs density as a determinant of NACI efficacy in cancers. Our scRNA-seq results indicated that the population of CD103+ CD8+ TILs was dramatically increased in the responders of NACI-treated HNSCC patients, while mIHC analysis confirmed the correlation between intratumoral CD103+ CD8+ TILs density and NACI efficacy in HNSCC patients. Further receiver operating characteristic curve analysis defined this TIL subset as a potent marker to predict patient response to NACI. Functional assays showed that CD103+ CD8+ TILs were tumor-reactive T cells, while programmed cell death protein-1 (PD-1) blockade enhanced CD103+ CD8+ TILs cytotoxicity against tumor growth in vivo. Mechanistically, targeting the triggering receptor expressed on myeloid cells 2-positive (TREM2+ ) macrophages might enhance the population of CD103+ CD8+ TILs and facilitate antitumor immunity during NACI treatment. CONCLUSIONS Our study highlights the impact of intratumoral CD103+ CD8+ TILs density on NACI efficacy in different cancers, while the efforts to elevate its population warrant further clinical investigation.
Collapse
Affiliation(s)
- Siqi Ren
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital of Sun Yat‐sen UniversityGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangdongP. R. China
| | - Tianjun Lan
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital of Sun Yat‐sen UniversityGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangdongP. R. China
| | - Fan Wu
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital of Sun Yat‐sen UniversityGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangdongP. R. China
| | - Suling Chen
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital of Sun Yat‐sen UniversityGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangdongP. R. China
| | - Xue Jiang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangdongP. R. China
| | - Chuying Huo
- Department of Gynecological OncologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangdongP. R. China
| | - Zitian Li
- School of Stomatology, Jilin UniversityJilinP. R. China
| | - Shule Xie
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital of Sun Yat‐sen UniversityGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangdongP. R. China
| | - Donghui Wu
- Stomatology Hospital of Haizhu districtGuangdongP. R. China
| | - Ruixin Wang
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital of Sun Yat‐sen UniversityGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangdongP. R. China
| | - Yanyan Li
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital of Sun Yat‐sen UniversityGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangdongP. R. China
| | - Lin Qiu
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital of Sun Yat‐sen UniversityGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangdongP. R. China
| | - Guoxin Huang
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital of Sun Yat‐sen UniversityGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangdongP. R. China
| | - Shurui Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangdongP. R. China
| | - Xiaojuan Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangdongP. R. China
| | - Meifeng Cen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangdongP. R. China
| | - Tingting Cai
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital of Sun Yat‐sen UniversityGuangdongP. R. China
| | - Zhaoyu Lin
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital of Sun Yat‐sen UniversityGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangdongP. R. China
| | - Jinsong Li
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital of Sun Yat‐sen UniversityGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangdongP. R. China
| | - Bowen Li
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital of Sun Yat‐sen UniversityGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangdongP. R. China
| |
Collapse
|
86
|
Oliveira G, Egloff AM, Afeyan AB, Wolff JO, Zeng Z, Chernock RD, Zhou L, Messier C, Lizotte P, Pfaff KL, Stromhaug K, Penter L, Haddad RI, Hanna GJ, Schoenfeld JD, Goguen LA, Annino DJ, Jo V, Oppelt P, Pipkorn P, Jackson R, Puram SV, Paniello RC, Rich JT, Webb J, Zevallos JP, Mansour M, Fu J, Dunn GP, Rodig SJ, Ley J, Morris LG, Dunn L, Paweletz CP, Kallogjeri D, Piccirillo JF, Adkins DR, Wu CJ, Uppaluri R. Preexisting tumor-resident T cells with cytotoxic potential associate with response to neoadjuvant anti-PD-1 in head and neck cancer. Sci Immunol 2023; 8:eadf4968. [PMID: 37683037 PMCID: PMC10794154 DOI: 10.1126/sciimmunol.adf4968] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 07/31/2023] [Indexed: 09/10/2023]
Abstract
About 50% of patients with locally advanced head and neck squamous cell carcinoma (HNSCC) experience recurrences after definitive therapy. The presurgical administration of anti-programmed cell death protein 1 (PD-1) immunotherapy results in substantial pathologic tumor responses (pTR) within the tumor microenvironment (TME). However, the mechanisms underlying the dynamics of antitumor T cells upon neoadjuvant PD-1 blockade remain unresolved, and approaches to increase pathologic responses are lacking. In a phase 2 trial (NCT02296684), we observed that 45% of patients treated with two doses of neoadjuvant pembrolizumab experienced marked pTRs (≥50%). Single-cell analysis of 17,158 CD8+ T cells from 14 tumor biopsies, including 6 matched pre-post neoadjuvant treatment, revealed that responding tumors had clonally expanded putative tumor-specific exhausted CD8+ tumor-infiltrating lymphocytes (TILs) with a tissue-resident memory program, characterized by high cytotoxic potential (CTX+) and ZNF683 expression, within the baseline TME. Pathologic responses after 5 weeks of PD-1 blockade were consistent with activation of preexisting CTX+ZNF683+CD8+ TILs, paralleling loss of viable tumor and associated tumor antigens. Response was associated with high numbers of CD103+PD-1+CD8+ T cells infiltrating pretreatment lesions, whereas revival of nonexhausted persisting clones and clonal replacement were modest. By contrast, nonresponder baseline TME exhibited a relative absence of ZNF683+CTX+ TILs and subsequent accumulation of highly exhausted clones. In HNSCC, revival of preexisting ZNF683+CTX+ TILs is a major mechanism of response in the immediate postneoadjuvant setting.
Collapse
Affiliation(s)
- Giacomo Oliveira
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School; Boston, MA, USA
| | - Ann Marie Egloff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School; Boston, MA, USA
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Alexander B. Afeyan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School; Boston, MA, USA
| | - Jacquelyn O. Wolff
- Center for Immuno-Oncology, Dana-Farber Cancer Institute; Boston, MA, USA
| | - Zexiang Zeng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rebecca D. Chernock
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Liye Zhou
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Cameron Messier
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute; Boston, MA, USA
| | - Patrick Lizotte
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute; Boston, MA, USA
| | - Kathleen L Pfaff
- Center for Immuno-Oncology, Dana-Farber Cancer Institute; Boston, MA, USA
| | - Kari Stromhaug
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Livius Penter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School; Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Hematology, Oncology and Tumor immunology, Campus Virchow Klinikum, Berlin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Robert I. Haddad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Glenn J. Hanna
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | | | - Laura A. Goguen
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Donald J. Annino
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Vickie Jo
- Department of Pathology, Brigham and Women’s Hospital; Boston, MA, USA
| | - Peter Oppelt
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine/Medical Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Patrik Pipkorn
- Department of Otolaryngology, Washington University School of Medicine; St. Louis, MO, USA
| | - Ryan Jackson
- Department of Otolaryngology, Washington University School of Medicine; St. Louis, MO, USA
| | - Sidharth V. Puram
- Department of Otolaryngology, Washington University School of Medicine; St. Louis, MO, USA
| | - Randal C. Paniello
- Department of Otolaryngology, Washington University School of Medicine; St. Louis, MO, USA
| | - Jason T. Rich
- Department of Otolaryngology, Washington University School of Medicine; St. Louis, MO, USA
| | - Jason Webb
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jose P. Zevallos
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mena Mansour
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Jingxin Fu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gavin P. Dunn
- Department of Neurological Surgery, Massachusetts General Hospital; Boston, MA, USA
| | - Scott J. Rodig
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital; Boston, MA, USA
| | - Jessica Ley
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine/Medical Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Luc G.T. Morris
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Lara Dunn
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Cloud P. Paweletz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute; Boston, MA, USA
| | - Dorina Kallogjeri
- Department of Otolaryngology, Washington University School of Medicine; St. Louis, MO, USA
| | - Jay F. Piccirillo
- Department of Otolaryngology, Washington University School of Medicine; St. Louis, MO, USA
| | - Douglas R. Adkins
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine/Medical Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School; Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Ravindra Uppaluri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School; Boston, MA, USA
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| |
Collapse
|
87
|
Chen S, He S, Wang D, Liu Y, Shao S, Tang L, Li C, Shi Q, Liu J, Wang F, Zhang S. Developing a predictive nomogram and web-based survival calculator for locally advanced hypopharyngeal cancer: A propensity score-adjusted, population-based study. BIOMOLECULES & BIOMEDICINE 2023; 23:902-913. [PMID: 37096424 PMCID: PMC10494849 DOI: 10.17305/bb.2023.8978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/06/2023] [Accepted: 04/06/2023] [Indexed: 04/26/2023]
Abstract
Understanding the clinical features and accurately predicting the prognosis of patients with locally advanced hypopharyngeal squamous cell carcinoma (LA-HPSCC) is important for patient centered decision-making. This study aimed to create a multi-factor nomogram predictive model and a web-based calculator to predict post-therapy survival for patients with LA-HPSCC. A retrospective cohort study analyzing Surveillance, Epidemiology, and End Results (SEER) database from 2004 to 2015 for patients diagnosed with LA-HPSCC was conducted and randomly divided into a training and a validation group (7:3 ratio). The external validation cohort included 276 patients from Sichuan Cancer Hospital, China. The Least Absolute Shrinkage and Selection Operator (LASSO)-Cox regression analysis was used to identify independent factors associated with overall survival (OS) and cancer-specific survival (CSS), and nomogram models and web-based survival calculators were constructed. Propensity score matching (PSM) was used to compare survival with different treatment options. A total of 2526 patients were included in the prognostic model. The median OS and CSS for the entire cohort were 20 (18.6-21.3) months and 24 (21.7-26.2) months, respectively. Nomogram models integrating the seven factors demonstrated high predictive accuracy for 3-year and 5-year survival. PSM found that patients who received surgery-based curative therapy had better OS and CSS than those who received radiotherapy-based treatment (median survival times: 33 months vs 18 months and 40 months vs 22 months, respectively). The nomogram model accurately predicted patient survival from LA-HPSCC. Surgery with adjuvant therapy yielded significantly better survival than definitive radiotherapy. and should be prioritized over definitive radiotherapy.
Collapse
Affiliation(s)
- Sihao Chen
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Center, Radiation Oncology Key Laboratory of Sichuan Province, Chengdu, China
| | - Shanshan He
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Center, Radiation Oncology Key Laboratory of Sichuan Province, Chengdu, China
| | - Dan Wang
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Center, Radiation Oncology Key Laboratory of Sichuan Province, Chengdu, China
| | - Yi Liu
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Center, Radiation Oncology Key Laboratory of Sichuan Province, Chengdu, China
| | - Shilong Shao
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Li Tang
- College of Public Health, Chongqing Medical University, Chongqing, China
| | - Chao Li
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Center, Radiation Oncology Key Laboratory of Sichuan Province, Chengdu, China
| | - Qiuling Shi
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Center, Radiation Oncology Key Laboratory of Sichuan Province, Chengdu, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Jifeng Liu
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Center, Radiation Oncology Key Laboratory of Sichuan Province, Chengdu, China
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Feng Wang
- Department of Medical Oncology, Cancer Center, West China Hospital, West China Medical School, Sichuan University, Sichuan, China
| | - Shichuan Zhang
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Center, Radiation Oncology Key Laboratory of Sichuan Province, Chengdu, China
| |
Collapse
|
88
|
van Dorp J, van der Heijden MS. The bladder cancer immune micro-environment in the context of response to immune checkpoint inhibition. Front Immunol 2023; 14:1235884. [PMID: 37727793 PMCID: PMC10505825 DOI: 10.3389/fimmu.2023.1235884] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/21/2023] [Indexed: 09/21/2023] Open
Abstract
Treatment with neoadjuvant cisplatin-based chemotherapy followed by radical cystectomy is the default treatment for muscle-invasive bladder cancer (BC). However, with the encouraging results of immune checkpoint inhibitiors (ICI) directed against PD-1/PD-L1 and CTLA-4 in recent years, the treatment landscape of BC is rapidly changing. In addition, it is becoming clear that the effect of ICI is highly dependent on the interaction between tumor cells and the tumor immune micro-environment (TIME). Different immune cells are involved in an anti-tumor response in BC. Cytotoxic CD8+ T-cells are the main effector cells, aided by other immune cells including other T-cells, B-cells and pro-inflammatory macrophages. As part of the ongoing anti-tumor immune response, lymphocytes aggregate in clusters called tertiary lymphoid structures (TLS). Tumor mutational burden (TMB) and infiltration of immune cells into the tumor are both important factors for establishing an anti-tumor immune response. In contrast, transforming growth factor beta (TGF-β) signaling in cancer-associated fibroblasts (CAFs) prevents infiltration of lymphocytes and potentially has an immunosuppressive effect. In conclusion, the effect of ICI seems to be reliant on a combination of tumor-intrinsic and TIME-related parameters. More research is needed to fully understand the underlying biological mechanisms to further improve patient care.
Collapse
Affiliation(s)
- Jeroen van Dorp
- Department of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Michiel S. van der Heijden
- Department of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
89
|
Stockem C, Mellema J, van Rhijn B, Boellaard T, van Montfoort M, Balduzzi S, Boormans J, Franckena M, Meijer R, Robbrecht D, Suelmann B, Schaake E, van der Heijden M. Induction therapy with ipilimumab and nivolumab followed by consolidative chemoradiation as organ-sparing treatment in urothelial bladder cancer: study protocol of the INDIBLADE trial. Front Oncol 2023; 13:1246603. [PMID: 37711193 PMCID: PMC10498281 DOI: 10.3389/fonc.2023.1246603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 07/31/2023] [Indexed: 09/16/2023] Open
Abstract
Introduction Studies that assessed the efficacy of pre-operative immune checkpoint blockade (ICB) in locally advanced urothelial cancer of the bladder showed encouraging pathological complete response rates, suggesting that a bladder-sparing approach may be a viable option in a subset of patients. Chemoradiation is an alternative for radical cystectomy with similar oncological outcomes, but is still mainly used in selected patients with organ-confined tumors or patients ineligible to undergo radical cystectomy. We propose to sequentially administer ICB and chemoradiation to patients with (locally advanced) muscle-invasive bladder cancer. Methods The INDIBLADE trial is an investigator-initiated, single-arm, multicenter phase 2 trial. Fifty patients with cT2-4aN0-2M0 urothelial bladder cancer will be treated with ipilimumab 3 mg/kg on day 1, ipilimumab 3 mg/kg plus nivolumab 1 mg/kg on day 22, and nivolumab 3 mg/kg on day 43 followed by chemoradiation. The primary endpoint is the bladder-intact event-free survival (BI-EFS). Events include: local or distant recurrence, salvage cystectomy, death and switch to platinum-based chemotherapy. We will also evaluate the potential of multiparametric magnetic resonance imaging of the bladder to identify non-responders, and we will assess the clearance of circulating tumor DNA as a biomarker for ICB treatment response. Discussion This is the first trial in which the efficacy of induction combination ICB followed by chemoradiation is being evaluated to provide bladder-preservation in patients with (locally advanced) urothelial bladder cancer. Clinical Trial Registration The INDIBLADE trial was registered on clinicaltrials.gov on January 21, 2022 (NCT05200988).
Collapse
Affiliation(s)
- C.F. Stockem
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - J.J.J. Mellema
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - B.W.G. van Rhijn
- Department of Oncological Urology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - T.N. Boellaard
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - M.L. van Montfoort
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - S. Balduzzi
- Department of Statistics, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - J.L. Boormans
- Department of Oncological Urology, Erasmus Medical Center, Rotterdam, Netherlands
| | - M. Franckena
- Department of Radiotherapy, Erasmus Medical Center, Rotterdam, Netherlands
| | - R.P. Meijer
- Department of Oncological Urology, University Medical Center (UMC), Utrecht, Netherlands
| | - D.G.J. Robbrecht
- Department of Medical Oncology, Erasmus Medical Center, Rotterdam, Netherlands
| | - B.B.M. Suelmann
- Department of Medical Oncology, University Medical Center (UMC), Utrecht, Netherlands
| | - E.E. Schaake
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - M.S. van der Heijden
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
90
|
Sun Q, Hong Z, Zhang C, Wang L, Han Z, Ma D. Immune checkpoint therapy for solid tumours: clinical dilemmas and future trends. Signal Transduct Target Ther 2023; 8:320. [PMID: 37635168 PMCID: PMC10460796 DOI: 10.1038/s41392-023-01522-4] [Citation(s) in RCA: 90] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/11/2023] [Accepted: 05/28/2023] [Indexed: 08/29/2023] Open
Abstract
Immune-checkpoint inhibitors (ICBs), in addition to targeting CTLA-4, PD-1, and PD-L1, novel targeting LAG-3 drugs have also been approved in clinical application. With the widespread use of the drug, we must deeply analyze the dilemma of the agents and seek a breakthrough in the treatment prospect. Over the past decades, these agents have demonstrated dramatic efficacy, especially in patients with melanoma and non-small cell lung cancer (NSCLC). Nonetheless, in the field of a broad concept of solid tumours, non-specific indications, inseparable immune response and side effects, unconfirmed progressive disease, and complex regulatory networks of immune resistance are four barriers that limit its widespread application. Fortunately, the successful clinical trials of novel ICB agents and combination therapies, the advent of the era of oncolytic virus gene editing, and the breakthrough of the technical barriers of mRNA vaccines and nano-delivery systems have made remarkable breakthroughs currently. In this review, we enumerate the mechanisms of each immune checkpoint targets, associations between ICB with tumour mutation burden, key immune regulatory or resistance signalling pathways, the specific clinical evidence of the efficacy of classical targets and new targets among different tumour types and put forward dialectical thoughts on drug safety. Finally, we discuss the importance of accurate triage of ICB based on recent advances in predictive biomarkers and diagnostic testing techniques.
Collapse
Affiliation(s)
- Qian Sun
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhenya Hong
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Cong Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Liangliang Wang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhiqiang Han
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Ding Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
91
|
Johnson JM, Vathiotis IA, Harshyne LA, Ali A, Bar Ad V, Axelrod R, Lorber E, Curry J, Cognetti DM, Luginbuhl AJ, Tuluc M, Keith S, Mahoney MG, Argiris A. Nivolumab and ipilimumab in combination with radiotherapy in patients with high-risk locally advanced squamous cell carcinoma of the head and neck. J Immunother Cancer 2023; 11:e007141. [PMID: 37536941 PMCID: PMC10401226 DOI: 10.1136/jitc-2023-007141] [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] [Accepted: 07/16/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND The combination of nivolumab and ipilimumab has been approved for the treatment of multiple solid tumors. This was a phase I study investigating definitive radioimmunotherapy (RIT) with nivolumab and ipilimumab for the treatment of locally advanced (LA) squamous cell carcinoma of the head and neck (SCCHN). METHODS Patients with newly diagnosed, stage IVA-IVB SCCHN eligible for cisplatin-based chemotherapy received nivolumab (3 mg/kg every 2 weeks for a total of 17 doses) and ipilimumab (1 mg/kg every 6 weeks for a total of 6 doses) starting 2 weeks prior to radiotherapy. The primary endpoint was safety of definitive RIT. Secondary endpoints included progression-free survival (PFS) and overall survival (OS). Exploratory endpoints included the association of baseline programmed death-ligand 1 (PD-L1) expression as well as on-treatment changes in immune bias with treatment outcomes. RESULTS Twenty-four patients were enrolled. With a median follow-up of 36.1 months, grade 3 or higher treatment-related adverse events were reported in 21 individuals (88%); 5 individuals developed in-field soft tissue ulceration during consolidation immunotherapy, resulting in one fatality. The 3-year PFS and OS rates were 74% (95% CI 58% to 94%) and 96% (95% CI 88% to 100%), respectively. PD-L1 combined positive score (CPS) did not correlate with death or disease progression. Decreases in extracellular vesicle PD-L1 within the concurrent RIT phase were associated with prolonged PFS (p=0.006). Also, interval decreases in circulating interleukin (IL)4, IL9, IL12, and IL17a during concurrent RIT were associated with subsequent ulceration. CONCLUSIONS Definitive RIT with nivolumab and ipilimumab has sufficient clinical activity to support further development. Early changes in circulating biomarkers appear able to predict treatment outcomes as well as ensuing in-field soft tissue ulceration. TRIAL REGISTRATION NUMBER NCT03162731.
Collapse
Affiliation(s)
- Jennifer M Johnson
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Otolaryngology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Ioannis A Vathiotis
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Larry A Harshyne
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ayesha Ali
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Voichita Bar Ad
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Rita Axelrod
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Emily Lorber
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Joseph Curry
- Department of Otolaryngology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - David M Cognetti
- Department of Otolaryngology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Adam J Luginbuhl
- Department of Otolaryngology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Madalina Tuluc
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Scott Keith
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Mỹ G Mahoney
- Department of Otolaryngology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Dermatology and Cuaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Athanassios Argiris
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
92
|
Giannini L, Alliata A, Cristofaro V, Incandela F, Pompilio M, Ottini A, Cavalieri S, Nuzzolese I, Iacovelli NA, Franceschini M, Deganello A. Radiation-Induced Oropharyngeal Squamous Cell Carcinoma: Case Report and Review of the Literature. Curr Oncol 2023; 30:6708-6719. [PMID: 37504352 PMCID: PMC10378216 DOI: 10.3390/curroncol30070492] [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: 05/31/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Radiation therapy (RT) is a mainstay for the treatment of head and neck (HN) cancers, with 80% of patients receiving such treatment. Radiation-induced malignancies represent a life-threatening long-term effect of RT, with an incidence of 0.5% to 15%. CASE DESCRIPTION After 13 years, a 33-year-old woman treated with chemo-radiotherapy for nasopharyngeal carcinoma developed a locally advanced, radiation-induced, p16-negative oropharyngeal squamous cell carcinoma (SCC) at the base of the tongue. Chemo/immunotherapy was administered as a first-line treatment. Given the optimal response and the feasibility of surgery, after three cycles, the patient underwent a total glossectomy, bilateral neck dissection, and reconstruction with a thoraco-dorsal free flap. A histological examination found SCC with a residual cancer burden of 70% and free margins. DISCUSSION The mechanisms responsible for carcinogenesis after RT are still not completely clear. Diagnosis may be challenging due to the previous treatment; growth patterns are unusual, and lymphotropism is lower. Prognosis is usually poor since surgical resectability is often not achievable. CONCLUSIONS Radiation-induced malignancies are difficult to treat. Patient management should always be discussed at a multidisciplinary level. Future research is needed to assess whether the promising results of clinical studies with pre-operative immunotherapy in locally advanced HN SCC patients may be translated into radiation-induced cancers.
Collapse
Affiliation(s)
- Lorenzo Giannini
- Department of Otorhinolaryngology, Maxillofacial, and Thyroid Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy
| | - Andrea Alliata
- Department of Otorhinolaryngology, Maxillofacial, and Thyroid Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Valentina Cristofaro
- Department of Otorhinolaryngology, Maxillofacial, and Thyroid Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Fabiola Incandela
- Department of Otorhinolaryngology, Maxillofacial, and Thyroid Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy
| | - Madia Pompilio
- Department of Otorhinolaryngology, Maxillofacial, and Thyroid Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy
| | - Arianna Ottini
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy
| | - Stefano Cavalieri
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Imperia Nuzzolese
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy
| | | | - Marzia Franceschini
- Radiotherapy Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy
| | - Alberto Deganello
- Department of Otorhinolaryngology, Maxillofacial, and Thyroid Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| |
Collapse
|
93
|
Sharon S, Daher-Ghanem N, Zaid D, Gough MJ, Kravchenko-Balasha N. The immunogenic radiation and new players in immunotherapy and targeted therapy for head and neck cancer. FRONTIERS IN ORAL HEALTH 2023; 4:1180869. [PMID: 37496754 PMCID: PMC10366623 DOI: 10.3389/froh.2023.1180869] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 06/27/2023] [Indexed: 07/28/2023] Open
Abstract
Although treatment modalities for head and neck cancer have evolved considerably over the past decades, survival rates have plateaued. The treatment options remained limited to definitive surgery, surgery followed by fractionated radiotherapy with optional chemotherapy, and a definitive combination of fractionated radiotherapy and chemotherapy. Lately, immunotherapy has been introduced as the fourth modality of treatment, mainly administered as a single checkpoint inhibitor for recurrent or metastatic disease. While other regimens and combinations of immunotherapy and targeted therapy are being tested in clinical trials, adapting the appropriate regimens to patients and predicting their outcomes have yet to reach the clinical setting. Radiotherapy is mainly regarded as a means to target cancer cells while minimizing the unwanted peripheral effect. Radiotherapy regimens and fractionation are designed to serve this purpose, while the systemic effect of radiation on the immune response is rarely considered a factor while designing treatment. To bridge this gap, this review will highlight the effect of radiotherapy on the tumor microenvironment locally, and the immune response systemically. We will review the methodology to identify potential targets for therapy in the tumor microenvironment and the scientific basis for combining targeted therapy and radiotherapy. We will describe a current experience in preclinical models to test these combinations and propose how challenges in this realm may be faced. We will review new players in targeted therapy and their utilization to drive immunogenic response against head and neck cancer. We will outline the factors contributing to head and neck cancer heterogeneity and their effect on the response to radiotherapy. We will review in-silico methods to decipher intertumoral and intratumoral heterogeneity and how these algorithms can predict treatment outcomes. We propose that (a) the sequence of surgery, radiotherapy, chemotherapy, and targeted therapy should be designed not only to annul cancer directly, but to prime the immune response. (b) Fractionation of radiotherapy and the extent of the irradiated field should facilitate systemic immunity to develop. (c) New players in targeted therapy should be evaluated in translational studies toward clinical trials. (d) Head and neck cancer treatment should be personalized according to patients and tumor-specific factors.
Collapse
Affiliation(s)
- Shay Sharon
- Department of Oral and Maxillofacial Surgery, Hadassah Medical Center, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Oral and Maxillofacial Surgery, Boston University and Boston Medical Center, Boston, MA, United States
| | - Narmeen Daher-Ghanem
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Deema Zaid
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michael J. Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States
| | - Nataly Kravchenko-Balasha
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
94
|
Hoffmann F, Franzen A, de Vos L, Wuest L, Kulcsár Z, Fietz S, Maas AP, Hollick S, Diop MY, Gabrielpillai J, Vogt T, Kuster P, Zarbl R, Dietrich J, Kristiansen G, Brossart P, Landsberg J, Strieth S, Dietrich D. CTLA4 DNA methylation is associated with CTLA-4 expression and predicts response to immunotherapy in head and neck squamous cell carcinoma. Clin Epigenetics 2023; 15:112. [PMID: 37415208 PMCID: PMC10327338 DOI: 10.1186/s13148-023-01525-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 06/25/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND The majority of patients with recurrent or metastasized head and neck squamous cell carcinoma (HNSCC) do not benefit from immune checkpoint blockade (ICB) while several patients experience severe and persistent immune-mediated side effects. Therefore, predictive biomarkers are urgently needed to allow for a personalized treatment. In this study, we investigated DNA methylation of the immune checkpoint gene CTLA4 with regard to its predictive value. METHODS We analyzed CTLA4 promoter methylation in tumors of HNSCC patients (N = 29) treated with ICB at the University Medical Center Bonn with regard to response to ICB and progression-free survival. We further analyzed a second cohort (N = 138) of patients that did not receive ICB with regard to CTLA4 promoter methylation, CTLA-4 protein expression, and immune cell infiltrates. Finally, we tested inducibility of CTLA-4 protein expression in HNSCC cells using the DNA methyltransferase inhibitor decitabine. RESULTS Lower CTLA4 promoter methylation correlated with response to ICB and prolonged progression-free survival. We could show that not only tumor infiltrating immune cells, but also HNSCC cells harbor cytoplasmic and nuclear CTLA-4 expression. CTLA4 promoter methylation inversely correlated with infiltrates of CD3+, CD4+, CD8+, and CD45+ immune cells. CTLA4 methylation did not correlate with protein expression in tumors, however, decitabine treatment led to decreased CTLA4 methylation and an induction of CTLA4 mRNA and CTLA-4 protein expression in HNSCC cell lines. CONCLUSIONS Our results indicate that CTLA4 DNA hypomethylation is a predictive biomarker for response to ICB in HNSCC. Our study warrants further analyses of the predictive value of CTLA4 DNA methylation in clinical trials of anti-PD-1 and/or anti-CTLA-4 immunotherapy in HNSCC.
Collapse
Grants
- 70113307 Deutsche Krebshilfe
- 2022-1A-08, 2020-1A-13, 2020-2A-06, 2022-4-18, 2023-4-06 University Medical Center Bonn BONFOR program
- 2022-1A-08, 2020-1A-13, 2020-2A-06, 2022-4-18, 2023-4-06 University Medical Center Bonn BONFOR program
- 2022-1A-08, 2020-1A-13, 2020-2A-06, 2022-4-18, 2023-4-06 University Medical Center Bonn BONFOR program
- 2022-1A-08, 2020-1A-13, 2020-2A-06, 2022-4-18, 2023-4-06 University Medical Center Bonn BONFOR program
- 2022-1A-08, 2020-1A-13, 2020-2A-06, 2022-4-18, 2023-4-06 University Medical Center Bonn BONFOR program
- Universitätsklinikum Bonn (8930)
Collapse
Affiliation(s)
- Friederike Hoffmann
- Department of Dermatology and Allergy, University Medical Center Bonn, Bonn, Germany
| | - Alina Franzen
- Department of Otorhinolaryngology, University Medical Center Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Luka de Vos
- Department of Dermatology and Allergy, University Medical Center Bonn, Bonn, Germany
- Department of Otorhinolaryngology, University Medical Center Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Lennert Wuest
- Department of Otorhinolaryngology, University Medical Center Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Zsófi Kulcsár
- Department of Otorhinolaryngology, University Medical Center Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Simon Fietz
- Department of Dermatology and Allergy, University Medical Center Bonn, Bonn, Germany
- Department of Otorhinolaryngology, University Medical Center Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Alexander Philippe Maas
- Department of Otorhinolaryngology, University Medical Center Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Sarah Hollick
- Department of Otorhinolaryngology, University Medical Center Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Marie Yatou Diop
- Department of Otorhinolaryngology, University Medical Center Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Jennis Gabrielpillai
- Department of Otorhinolaryngology, University Medical Center Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Timo Vogt
- Department of Otorhinolaryngology, University Medical Center Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Pia Kuster
- Department of Dermatology and Allergy, University Medical Center Bonn, Bonn, Germany
| | - Romina Zarbl
- Department of Otorhinolaryngology, University Medical Center Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Joern Dietrich
- Department of Otorhinolaryngology, University Medical Center Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Glen Kristiansen
- Institute of Pathology, University Medical Center Bonn, Bonn, Germany
| | - Peter Brossart
- Department of Oncology, Hematology and Rheumatology, University Medical Center Bonn, Bonn, Germany
| | - Jennifer Landsberg
- Department of Dermatology and Allergy, University Medical Center Bonn, Bonn, Germany
| | - Sebastian Strieth
- Department of Otorhinolaryngology, University Medical Center Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Dimo Dietrich
- Department of Otorhinolaryngology, University Medical Center Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
95
|
Holzgreve A, Taugner J, Käsmann L, Müller P, Tufman A, Reinmuth N, Li M, Winkelmann M, Unterrainer LM, Nieto AE, Bartenstein P, Kunz WG, Ricke J, Belka C, Eze C, Unterrainer M, Manapov F. Metabolic patterns on [ 18F]FDG PET/CT in patients with unresectable stage III NSCLC undergoing chemoradiotherapy ± durvalumab maintenance treatment. Eur J Nucl Med Mol Imaging 2023; 50:2466-2476. [PMID: 36951991 PMCID: PMC10250493 DOI: 10.1007/s00259-023-06192-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/05/2023] [Indexed: 03/24/2023]
Abstract
PURPOSE In patients with unresectable stage III non-small-cell lung cancer (NSCLC), durvalumab maintenance treatment after chemoradiotherapy (CRT) significantly improves survival. So far, however, metabolic changes of tumoral lesions and secondary lymphoid organs under durvalumab are unknown. Hence, we assessed changes on [18F]FDG PET/CT in comparison to patients undergoing CRT alone. METHODS Forty-three patients with [18F]FDG PET/CT both before and after standard CRT for unresectable stage III NSCLC were included, in 16/43 patients durvalumab maintenance treatment was initiated (CRT-IO) prior to the second PET/CT. Uptake of tumor sites and secondary lymphoid organs was compared between CRT and CRT-IO. Also, readers were blinded for durvalumab administration and reviewed scans for findings suspicious for immunotherapy-related adverse events (irAE). RESULTS Initial uptake characteristics were comparable. However, under durvalumab, diverging metabolic patterns were noted: There was a significantly higher reduction of tumoral uptake intensity in CRT-IO compared to CRT, e.g. median decrease of SUVmax -70.0% vs. -24.8%, p = 0.009. In contrast, the spleen uptake increased in CRT-IO while it dropped in CRT (median + 12.5% vs. -4.4%, p = 0.029). Overall survival was significantly longer in CRT-IO compared to CRT with few events (progression/death) noted in CRT-IO. Findings suggestive of irAE were present on PET/CT more often in CRT-IO (12/16) compared to CRT (8/27 patients), p = 0.005. CONCLUSION Durvalumab maintenance treatment after CRT leads to diverging tumoral metabolic changes, but also increases splenic metabolism and leads to a higher proportion of findings suggestive of irAE compared to patients without durvalumab. Due to significantly prolonged survival with durvalumab, survival analysis will be substantiated in correlation to metabolic changes as soon as more clinical events are present.
Collapse
Affiliation(s)
- Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
| | - Julian Taugner
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Lukas Käsmann
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
- Member of the German Center for Lung Research (DZL), Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Philipp Müller
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Amanda Tufman
- Member of the German Center for Lung Research (DZL), Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany
- Department of Internal Medicine V, University Hospital, LMU Munich, Munich, Germany
| | | | - Minglun Li
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Michael Winkelmann
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Lena M Unterrainer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Alexander E Nieto
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Wolfgang G Kunz
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
- Member of the German Center for Lung Research (DZL), Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Chukwuka Eze
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Farkhad Manapov
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
- Member of the German Center for Lung Research (DZL), Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| |
Collapse
|
96
|
Mortezaee K, Majidpoor J, Kharazinejad E. The impact of hypoxia on tumor-mediated bypassing anti-PD-(L)1 therapy. Biomed Pharmacother 2023; 162:114646. [PMID: 37011483 DOI: 10.1016/j.biopha.2023.114646] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/04/2023] Open
Abstract
Extending the durability of response is the current focus in cancer immunotherapy with immune checkpoint inhibitors (ICIs). However, factors like non-immunogenic tumor microenvironment (TME) along with aberrant angiogenesis and dysregulated metabolic systems are negative contributors. Hypoxia is a key TME condition and a critical promoter of tumor hallmarks. It acts on immune and non-immune cells within TME in order for promoting immune evasion and therapy resistance. Extreme hypoxia is a major promoter of resistance to the programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) inhibitor therapy. Hypoxia inducible factor-1 (HIF-1) acts as a key mediator of hypoxia and a critical promoter of resistance to the anti-PD-(L)1. Targeting hypoxia or HIF-1 can thus be an effective strategy for reinvigoration of cellular immunity against cancer. Among various strategies presented so far, the key focus is over vascular normalization, which is an approach highly effective for reducing the rate of hypoxia, increasing drug delivery into the tumor area, and boosting the efficacy of anti-PD-(L)1.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Islamic Republic of Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Islamic Republic of Iran
| | - Ebrahim Kharazinejad
- Department of Anatomy, Faculty of Medicine, Abadan University of Medical Sciences, Abadan, Islamic Republic of Iran.
| |
Collapse
|
97
|
Benavente S. Remodeling the tumor microenvironment to overcome treatment resistance in HPV-negative head and neck cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:291-313. [PMID: 37457128 PMCID: PMC10344731 DOI: 10.20517/cdr.2022.141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 05/02/2023] [Accepted: 05/22/2023] [Indexed: 07/18/2023]
Abstract
Despite intensive efforts and refined techniques, overall survival in HPV-negative head and neck cancer remains poor. Robust immune priming is required to elicit a strong and durable antitumor immune response in immunologically cold and excluded tumors like HPV-negative head and neck cancer. This review highlights how the tumor microenvironment could be affected by different immune and stromal cell types, weighs the need to integrate metabolic regulation of the tumor microenvironment into cancer treatment strategies and summarizes the emerging clinical applicability of personalized immunotherapeutic strategies in HPV-negative head and neck cancer.
Collapse
Affiliation(s)
- Sergi Benavente
- Correspondence to: Dr. Sergi Benavente, Department of Radiation Oncology, Vall d’Hebron University Hospital, Passeig Vall d’Hebron 119, Barcelona 08035, Spain. E-mail:
| |
Collapse
|
98
|
Taverna C, Franchi A. Role of Surgical Pathologist for Detection of Immunooncologic Predictive Factors in Head and Neck Cancer. Adv Anat Pathol 2023; 30:167-173. [PMID: 36175939 DOI: 10.1097/pap.0000000000000374] [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: 11/27/2022]
Abstract
Immunotherapy has shown promising results in the treatment of recurrent and metastatic head and neck cancers. Antiprogrammed cell death (PD)-1 therapies have been recently approved in this setting and they are currently tested also in the treatment of locally advanced diseases and in the neoadjuvant setting. However, the clinical benefits of these treatments have been quite variable, hence the need to select those patients who may obtain the maximal efficacy through the identification of predictive biomarkers. Currently, PD-L1 immunohistochemical expression by tumor and immune cells is the most widely used predictive biomarker for immunotherapy in head and neck squamous cell carcinoma. Nevertheless, patients with PD-L1 - tumors may still respond to treatments, thereby emphasizing the need for the identification of other predictive biomarkers. In this review, we summarize the current data on histologic and molecular parameters that can be used to select patients with head and neck cancers for immunotherapy, with a focus on squamous cell carcinoma and salivary gland carcinomas.
Collapse
Affiliation(s)
- Cecilia Taverna
- Department of Translational Research and of New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | |
Collapse
|
99
|
Marret G, Borcoman E, Le Tourneau C. Window-of-opportunity clinical trials for biomarker discovery in head and neck squamous cell carcinoma. Curr Opin Oncol 2023; 35:158-165. [PMID: 36966501 DOI: 10.1097/cco.0000000000000940] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2023]
Abstract
PURPOSE OF REVIEW We review the window-of-opportunity clinical trials that have been reported in head and neck squamous cell carcinoma (HNSCC), and discuss their challenges. RECENT FINDINGS Limited treatment options exist in HNSCC. Cetuximab, an mAb targeting epidermal growth factor receptor, and the PD-1 inhibitors nivolumab and pembrolizumab, are the only drugs that improved overall survival in the recurrent and/or metastatic setting. Both cetuximab and nivolumab improve overall survival by less than 3 months, potentially because of the lack of predictive biomarkers. The only validated predictive biomarker to date is protein ligand PD-L1 expression that predicts the efficacy of pembrolizumab in first-line, nonplatinum refractory recurrent and/or metastatic HNSCC. The identification of biomarkers of efficacy of new drugs is key to avoid administering toxic drugs to patients who will not benefit from them, and to expect increased drug efficacy in the biomarker-positive group of patients. One way of identifying such biomarkers are the window-of-opportunity trials in which drugs are given for a short period of time before the definitive treatment, with the aim to collect samples for translational research. These trials differ from neoadjuvant strategies where efficacy is the primary endpoint. SUMMARY We show that these trials were safe and successful in identifying biomarkers.
Collapse
Affiliation(s)
- Grégoire Marret
- Department of Drug Development and Innovation (D3i), Institut Curie
| | - Edith Borcoman
- Department of Drug Development and Innovation (D3i), Institut Curie
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie
- INSERM U900, Institut Curie, Paris-Saclay University, Paris, France
| |
Collapse
|
100
|
Preissner S, Heiland M, Preissner R, Wirth M, Wollenberg B. Antibiotics Significantly Decrease the Survival of Head and Neck Carcinoma Patients with Immunotherapy: A Real-World Analysis of More Than 3000 Cases. Cancers (Basel) 2023; 15:cancers15082342. [PMID: 37190270 DOI: 10.3390/cancers15082342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/09/2023] [Accepted: 04/15/2023] [Indexed: 05/17/2023] Open
Abstract
OBJECTIVE The human gut microbiome is strongly influenced by the administration of drugs, namely antibiotics. We hypothesized that the effectiveness of immunotherapy with pembrolizumab in oral squamous cell carcinoma patients is decreased by the administration of antibiotics three months before and after immunotherapy. METHODS We retrieved data from patients diagnosed with head and neck squamous cell carcinoma (HNSCC) (International Classification of Diseases [ICD]-10 codes C00-C14) and receiving immunotherapy with pembrolizumab from the TriNetX network. Two cohorts were built: patients in cohort I did not receive any antibiotics within three months before or up to three months after immunotherapy, while patients in cohort II were administered antibiotics at least once within three months before or after immunotherapy. To exclude confounders, we matched cohorts 1:1 for age, sex, secondary lymph node metastases, nicotine dependence, the insertion of feeding devices, body mass index (BMI) and severe sepsis. After defining the primary outcome as "death", a Kaplan-Meier analysis was performed, and the risk ratio (RR), odds ratio (OR) and hazard ratio (HR) were calculated. RESULTS A total of 3651 patients were enrolled, and after matching, each cohort consisted of 1362 patients. Among cohorts I and II, 346 and 511 patients were deceased within one year (risk of death = 25.5 and 38.3%, respectively), whereby the risk difference was significant (p = 0.000; log-rank test). The RR was 0.68 (95% confidence interval: 0.60-0.76), OR was 0.57 (0.48-0.67) and HR was 0.58 (0.51-0.67). CONCLUSIONS Our hypothesis was confirmed: administering antibiotics significantly decreases the drug effectiveness of immunotherapy. We hypothesize that this finding is associated with antibiotic-related changes in the gut microbiome. Prospective clinical studies on the gut microbiome in cancer patients are necessary to understand the complex ecosystem of microbiota during immunotherapy. TRIAL REGISTRATION Due to the retrospective nature of the study, no registration was necessary.
Collapse
Affiliation(s)
- Saskia Preissner
- Department of Oral and Maxillofacial Surgery, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Max Heiland
- Department of Oral and Maxillofacial Surgery, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Robert Preissner
- Institute of Physiology and Science-IT, Charité-Universitätsmedizin Berlin, Philippstr. 12, 10115 Berlin, Germany
| | - Markus Wirth
- Clinic for Otorhinolaryngology, Head and Neck Surgery, Klinikum Rechts der Isar der Technischen Universität München (MRI TUM), Ismaningerstr. 22, 81675 Munich, Germany
| | - Barbara Wollenberg
- Clinic for Otorhinolaryngology, Head and Neck Surgery, Klinikum Rechts der Isar der Technischen Universität München (MRI TUM), Ismaningerstr. 22, 81675 Munich, Germany
| |
Collapse
|