1
|
Talebi F, Gregucci F, Ahmed J, Ben Chetrit N, D. Brown B, Chan TA, Chand D, Constanzo J, Demaria S, I. Gabrilovich D, Golden E, Godkin A, Guha C, P. Gupta G, Hasan A, G. Herrera F, Kaufman H, Li D, A. Melcher A, McDonald S, Merghoub T, Monjazeb AM, Paris S, Pitroda S, Sadanandam A, Schaue D, Santambrogio L, Szapary P, Sage J, W. Welsh J, Wilkins A, H. Young K, Wennerberg E, Zitvogel L, Galluzzi L, Deutsch E, C. Formenti S. Updates on radiotherapy-immunotherapy combinations: Proceedings of 8th Annual ImmunoRad Conference. Oncoimmunology 2025; 14:2507856. [PMID: 40401900 PMCID: PMC12101595 DOI: 10.1080/2162402x.2025.2507856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/23/2025] Open
Abstract
The annual ImmunoRad Conference has established itself as a recurrent occasion to explore the possibility of combining radiation therapy (RT) and immunotherapy (IT) for clinical cancer management. Bringing together a number of preclinical and clinical leaders in the fields of radiation oncology, immuno-oncology and IT, this annual event fosters indeed essential conversations and fruitful exchanges on how to address existing challenges to expand the therapeutic value of RT-IT combinations. The 8th edition of the ImmunoRad Conference, which has been held in October 2024 at the Weill Cornell Medical College of New York City, highlighted exciting preclinical and clinical advances at the interface between RT and IT, setting the stage for extra progress toward extended benefits for patients with an increasing variety of tumor types. Here, we critically summarize the lines of investigation that have been discussed at the occasion of the 8th Annual ImmunoRad Conference.
Collapse
Affiliation(s)
- Fereshteh Talebi
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Fabiana Gregucci
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Jalal Ahmed
- Icahn Genomics Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nir Ben Chetrit
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Brian D. Brown
- Icahn Genomics Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Timothy A. Chan
- Department of Cancer Sciences, Global Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Case Western University School of Medicine, Cleveland, OH, USA
| | | | - Julie Constanzo
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | | | - Encouse Golden
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Andrew Godkin
- Division of Infection and Immunity/Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff, UK
| | - Chandan Guha
- Departments of Radiation Oncology and Pathology, Albert Einstein College of Medicine, New York, NY, USA
| | - Gaorav P. Gupta
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Fernanda G. Herrera
- AGORA Cancer Research Center, Swiss Cancer Center Leman, Lausanne, Switzerland
- Services of Radiation Oncology and Immuno-Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- Department of Oncology, Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland
| | | | - Donna Li
- University of Wisconsin, Madison, WI, USA
| | - Alan A. Melcher
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Sierra McDonald
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center and Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, NY, USA
| | - Arta M. Monjazeb
- Department of Radiation Oncology, University of California, San Diego, CA, USA
| | | | - Sean Pitroda
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Anguraj Sadanandam
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | | | - Julien Sage
- Departments of Genetics and Pediatrics, Stanford University, Stanford, California
| | - James W. Welsh
- Department of Radiation Oncology, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Anna Wilkins
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Kristina H. Young
- Division of Radiation Oncology, The Oregon Clinic, Portland, OR, USA
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Eric Wennerberg
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Laurence Zitvogel
- Gustave Roussy, INSERM U1015, Division of Medicine, Paris-Saclay University, Center of Clinical Investigations BIOTHERIS, Villejuif, France
| | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy, INSERM U1030, Division of Medicine, Paris-Saclay University, RHU LySAIRI “Lymphocyte-Sparing Artificial Intelligence-guided Radio-Immunotherapy”, Villejuif, France
| | - Silvia C. Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
2
|
Wu KP, Li QQ, Luo XQ, Wang XX, Lai YZ, Tian D, Yang HC, Wei XL, Wang LY, Li QM, Zhu D, Chen SJ, Li YS. Chemoimmunotherapy as induction treatment in concurrent chemoradiotherapy for patients with nasopharyngeal carcinoma stage IVa. Ann Med 2025; 57:2453091. [PMID: 39834281 PMCID: PMC11753007 DOI: 10.1080/07853890.2025.2453091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/11/2024] [Accepted: 12/29/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Chemoimmunotherapy is the first-line therapy for patients with recurrent or metastatic nasopharyngeal carcinoma (NPC) and is currently the main induction treatment option for patients with locoregionally advanced NPC. However, it remains unclear whether combining immunotherapy with standard induction chemotherapy enhances its efficacy. This study aimed to evaluate the efficacy, toxicity, and survival outcomes of induction chemoimmunotherapy in patients with locoregionally advanced NPC. METHODS This study analyzed 50 patients with stage IVa NPC between January 2020 and December 2023 in our hospital. Among them, 23 received induction chemoimmunotherapy, and 27 received induction chemotherapy. All patients underwent standard platinum-based concurrent intensity-modulated radiation therapy. We compared tumor response and toxicity during induction treatment and concurrent chemoradiotherapy (CCRT) between the two groups. RESULTS The objective and complete response rates were significantly higher in the induction chemoimmunotherapy group compared to the induction chemotherapy group (95.7% vs 77.8%, and 39.1% vs 22.2%, respectively). All patients completed radical CCRT. Median follow-up was 24 months. Patients who received induction chemoimmunotherapy had longer event-free survival (EFS) compared to those who received induction chemotherapy (p = 0.029, Hazard Ratio and 95%confidence interval [CI]: 0.24 [0.07-0.85]). The 24-month EFS was higher in the induction chemoimmunotherapy group compared with the chemotherapy group (24-month EFS rates and 95%CI: 88.9% [95%CI: 68.3%-100%] vs 62.6% [95%CI: 43.1%-82.1%]). No significant differences in adverse events were observed between the two groups during induction treatment and CCRT. CONCLUSIONS Adding immunotherapy to induction chemotherapy may be an effective and safe choice for treating patients with stage IVa NPC.
Collapse
Affiliation(s)
- Kun-Peng Wu
- Department of Oncology, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
- Heyuan Key Laboratory of Molecular Diagnosis & Disease Prevention and Treatment, Doctors Station of Guangdong Province, Heyuan People’s Hospital, Heyuan, Guangdong, China
| | - Qing-Qing Li
- Department of Oncology, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
- Heyuan Key Laboratory of Molecular Diagnosis & Disease Prevention and Treatment, Doctors Station of Guangdong Province, Heyuan People’s Hospital, Heyuan, Guangdong, China
| | - Xu-Qiang Luo
- Department of Oncology, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
- Heyuan Key Laboratory of Molecular Diagnosis & Disease Prevention and Treatment, Doctors Station of Guangdong Province, Heyuan People’s Hospital, Heyuan, Guangdong, China
| | - Xiao-Xi Wang
- Department of Otolaryngology-Head and Neck Surgery, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yan-Zhen Lai
- Department of Oncology, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
- Heyuan Key Laboratory of Molecular Diagnosis & Disease Prevention and Treatment, Doctors Station of Guangdong Province, Heyuan People’s Hospital, Heyuan, Guangdong, China
| | - Dan Tian
- Department of Oncology, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
- Heyuan Key Laboratory of Molecular Diagnosis & Disease Prevention and Treatment, Doctors Station of Guangdong Province, Heyuan People’s Hospital, Heyuan, Guangdong, China
| | - Hong-cheng Yang
- Department of Oncology, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
- Heyuan Key Laboratory of Molecular Diagnosis & Disease Prevention and Treatment, Doctors Station of Guangdong Province, Heyuan People’s Hospital, Heyuan, Guangdong, China
| | - Xue-Ling Wei
- Department of Oncology, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
| | - Lan-Ying Wang
- Department of Oncology, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
| | - Qiu-Ming Li
- Department of Otolaryngology-Head and Neck Surgery, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
| | - Dao Zhu
- Department of Otolaryngology-Head and Neck Surgery, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
| | - Si-Jie Chen
- Department of Otolaryngology-Head and Neck Surgery, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
| | - Yang-Si Li
- Department of Oncology, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
- Heyuan Key Laboratory of Molecular Diagnosis & Disease Prevention and Treatment, Doctors Station of Guangdong Province, Heyuan People’s Hospital, Heyuan, Guangdong, China
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
3
|
Morgan D, Okwuone DD, Berggren KL, Arnold L, Schmidt A, Spiess C, Smith H, Yada R, Hendrikse N, Madan R, Shrock D, Lominska C, Hu M, Witek M, Soper S, Lin Y, Gao H, McCance DJ, Thomas SM, Beebe D, Kerr SC, Gan GN. MK2 promotes p16 negative head and neck cancer migration, invasion, and metastasis. Cancer Lett 2025; 622:217690. [PMID: 40185303 DOI: 10.1016/j.canlet.2025.217690] [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: 02/03/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
For patients with locally advanced, p16-negative head and neck squamous cell carcinoma (HNSCC), overall survival remains poor due to primary locoregional failure and distant metastasis following curative therapy. We aimed to understand how MAPKAPK2 (MK2) regulates HNSCC tumor cell migration and invasion, important first steps in cancer metastases. The TCGA database and HNSCC tissue microarrays were used to show that MK2 expression was associated with more advanced cancers and faster cancer recurrence rates. We observed that silencing of tumor MK2 in human cell lines (shRNA) caused a significant reduction in tumor cell migration-invasion in a complex HNSCC microphysiologic system used to recapitulate the tumor microenvironment. Murine cells (Ly2) with MK2 silenced (CRISPR-Cas9) also demonstrated reduced migration and invasion using 2D and 3D monoculture cell migration-invasions assays. Ly2 cells are orthotopic p16-negative murine metastatic cells that spontaneously metastasize, and we observed that MK2 inhibition via genetic (Cas9/CRISPR) or pharmacologic (PF-3644022) methods led to a significant reduction in the number of circulating tumor cells, fewer lymph node and lung metastases, and MK2 inhibited mice showed improved overall survival. Our findings suggest that HNSCC MK2 regulates tumor cell migration-invasion and may be a promising therapeutic target to reduce metastases.
Collapse
Affiliation(s)
- Deri Morgan
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Dakota Dd Okwuone
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | | | - Levi Arnold
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Alyssa Schmidt
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Colby Spiess
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Hannah Smith
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ravi Yada
- Carbone Cancer Center, University of Wisconsin Madison, WI, USA
| | | | - Rashna Madan
- Department of Pathology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Devin Shrock
- Department of Pathology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Chris Lominska
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Mengjia Hu
- Department of Chemistry, University of Kansas, Topeka, KS, USA
| | | | - Steven Soper
- Department of Chemistry, University of Kansas, Topeka, KS, USA
| | - Yuting Lin
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Hao Gao
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Dennis J McCance
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sufi M Thomas
- Department of Pathology, The University of New Mexico, Albuquerque, NM, USA
| | - David Beebe
- Carbone Cancer Center, University of Wisconsin Madison, WI, USA
| | - Sheena C Kerr
- Carbone Cancer Center, University of Wisconsin Madison, WI, USA
| | - Gregory N Gan
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
4
|
Oliveira Dias J, Sampaio Fagundes I, Bisio MDC, da Silva Barboza V, Jacinto AA, Altei WF. Extracellular vesicles as the common denominator among the 7 Rs of radiobiology: From the cellular level to clinical practice. Biochim Biophys Acta Rev Cancer 2025; 1880:189315. [PMID: 40216093 DOI: 10.1016/j.bbcan.2025.189315] [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: 11/22/2024] [Revised: 04/03/2025] [Accepted: 04/03/2025] [Indexed: 04/17/2025]
Abstract
Extracellular vesicles (EVs) are lipid-bound particles released by tumor cells and widely explored in cancer development, progression, and treatment response, being considered as valuable components to be explored as biomarkers or cellular targets to modulate the effect of therapies. The mechanisms underlying the production and profile of EVs during radiotherapy (RT) require addressing radiobiological aspects to determine cellular responses to specific radiation doses and fractionation. In this review, we explore the role of EVs in the 7 Rs of radiobiology, known as the molecular basis of a biological tissue response to radiation, supporting EVs as a shared player in all the seven processes. We also highlight the relevance of EVs in the context of liquid biopsy and resistance to immunotherapy, aiming to establish the connection and utility of EVs as tools in contemporary and precision radiotherapy.
Collapse
Affiliation(s)
- Júlia Oliveira Dias
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | | | | | | | | | - Wanessa Fernanda Altei
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil; Radiation Oncology Department, Barretos Cancer Hospital, Barretos, Brazil.
| |
Collapse
|
5
|
Pisljar Z, Markelc B, Brezar SK, Bozic T, Sersa G, Cemazar M, Jesenko T. Partial versus whole tumor-volume irradiation in combination with immunotherapy: Comparable outcomes in immunosuppressed mouse models of oral squamous cell carcinoma. Biomed Pharmacother 2025; 187:118107. [PMID: 40288172 DOI: 10.1016/j.biopha.2025.118107] [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: 03/04/2025] [Revised: 04/01/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025] Open
Abstract
Radiotherapy is a standard therapy for oral squamous cell carcinoma (OSCC) with immunomodulatory potential. Due to high lymphocyte radiosensitivity, partial tumor-volume irradiation (pIR), targeting only part of the tumor, is being explored for immunomodulation. This study compared the effects of whole tumor-volume irradiation (IR) and pIR, targeting approximately 50 % of the tumor volume, in combination with anti-PD-1 immune checkpoint inhibitors (ICI). The therapeutic efficacy of a single 15 Gy IR or pIR dose combined with anti-PD-1 ICI was evaluated in two immune cold murine OSCC models: human papillomavirus (HPV)-negative MOC1 and HPV-positive MOC1-HPV K1 stably expressing HPV-16 oncogenes E6/E7. Additionally, immune cell populations in the tumor microenvironment (TME) were analyzed using flow cytometry. Both IR and pIR induced transient immune cell infiltration in the TME. However, pIR led to significantly lower tumor growth inhibition than IR. While IR + ICI failed to improve survival compared to IR alone, pIR + ICI significantly prolonged survival compared to pIR alone in the MOC1 model, along with increase in cytotoxic T cell infiltration. In the MOC1-HPV K1 model, responses varied. Responding tumors were enriched with effector memory T cells, whereas non-responders exhibited increased neutrophil (MDSCs) and monocyte-derived dendritic cells infiltration. The study indicates that while pIR has immunomodulatory potential, its effects are comparable to IR in the tested settings. Further research is needed to optimize dosing and scheduling for pIR and anti-PD-1 ICI. Additionally, combination with other immunotherapies could be explored in further studies to enhance treatment efficacy in immune cold OSCC models.
Collapse
Affiliation(s)
- Ziva Pisljar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, Ljubljana, Slovenia; University of Ljubljana, Faculty of Medicine, Vrazov trg 2, Ljubljana, Slovenia
| | - Bostjan Markelc
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, Ljubljana, Slovenia; University of Ljubljana, Biotechnical Faculty, Jamnikarjeva ulica 101, Ljubljana, Slovenia
| | - Simona Kranjc Brezar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, Ljubljana, Slovenia; University of Ljubljana, Faculty of Medicine, Vrazov trg 2, Ljubljana, Slovenia
| | - Tim Bozic
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, Ljubljana, Slovenia
| | - Gregor Sersa
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, Ljubljana, Slovenia; University of Ljubljana, Faculty of Health Sciences, Zdravstvena pot 5, Ljubljana, Slovenia
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, Ljubljana, Slovenia; University of Primorska, Faculty of Education, Cankarjeva pot 5, Koper, Slovenia.
| | - Tanja Jesenko
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, Ljubljana, Slovenia; University of Ljubljana, Faculty of Medicine, Vrazov trg 2, Ljubljana, Slovenia.
| |
Collapse
|
6
|
Zandberg DP, Vujanovic L, Clump DA, Isett BP, Wang H, Sica G, Bao R, Li H, Ohr J, Skinner HD, Seethala RR, Chiosea SI, Ferris RL, Bauman JE. Randomized Phase II Study of Concurrent Versus Sequential Pembrolizumab in Combination With Chemoradiation in Locally Advanced Head and Neck Cancer. J Clin Oncol 2025:JCO2401580. [PMID: 40424564 DOI: 10.1200/jco-24-01580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 01/16/2025] [Accepted: 03/28/2025] [Indexed: 05/29/2025] Open
Abstract
PURPOSE The optimal timing of pembrolizumab with chemoradiation (CRT) in locally advanced (LA) head and neck squamous cell carcinoma (HNSCC) is unknown. PATIENTS AND METHODS Our phase II trial randomly assigned patients 1:1 to concurrent pembrolizumab (200 mg once every 3 weeks × 8) starting 1 week before CRT (cisplatin 40 mg/m2 once weekly + radiation 70 Gy) versus sequential pembrolizumab starting 2 weeks after CRT. Human papillomavirus (HPV)+ (>10 pack-years or T4 or N3) and HPV(-) LA HNSCC were included, stratified by HPV and N stage. In our pick-the-winner design, if both arms met the trivariate primary end point (1-year locoregional failure <60%, progression-free survival [PFS] ≥60%, and dose limiting toxicity rate ≤20%), the arm with numerically superior 1-year PFS would be selected. Survival end points were compared by a univariate Cox model. Pretreatment and on-treatment tumor biopsies (week 2 of CRT) were evaluated by multispectral imaging and compared using two-sided paired t-tests. RESULTS Treated patients (41 concurrent and 39 sequential) were 71% oropharynx (53% HPV+), 92.5% stage IV (46% T4, 76% N2), similar by arm. Both arms met the trivariate primary end point, with superior 1-year PFS in the sequential arm (84% v 71%) and favorable 4-year outcomes: locoregional control (96% v 64%; hazard ratio [HR], 0.11 [95% CI, 0.01 to 0.89]; P = .012), PFS (69% v 49%; HR, 0.55 [95% CI, 0.25 to 1.22]; P = .132), and overall survival (83% v 71%; HR, 0.51 [95% CI, 0.19 to 1.37]; P = .17). There was a significant increase in macrophages, PD-L1+ macrophages, and PD-L1+ tumor cells with treatment in the concurrent but not the sequential arm. CONCLUSION CRT with sequential pembrolizumab met criteria for further study. Immunosuppressive changes in the TME differed between arms, reflecting the impact of one dose of pembrolizumab in the concurrent arm.
Collapse
Affiliation(s)
- Dan P Zandberg
- UPMC Hillman Cancer Center, Pittsburgh, PA
- Division of Hematology/Oncology, University of Pittsburgh, Pittsburgh, PA
| | - Lazar Vujanovic
- UPMC Hillman Cancer Center, Pittsburgh, PA
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA
| | - David A Clump
- Department of Radiation Oncology, West Virginia University, Morgantown, WV
| | - Brian P Isett
- UPMC Hillman Cancer Center, Pittsburgh, PA
- UPMC Hillman Cancer Center Bioinformatics Core, Pittsburgh, PA
| | - Hong Wang
- UPMC Hillman Cancer Center, Pittsburgh, PA
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA
| | - Gabriel Sica
- UPMC Hillman Cancer Center, Pittsburgh, PA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - Riyue Bao
- UPMC Hillman Cancer Center, Pittsburgh, PA
- UPMC Hillman Cancer Center Bioinformatics Core, Pittsburgh, PA
| | - Housayin Li
- Molecular Genetics and Developmental Biology Graduate Program, University of Pittsburgh, Pittsburgh, PA
| | - James Ohr
- UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Heath D Skinner
- UPMC Hillman Cancer Center, Pittsburgh, PA
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA
| | - Raja R Seethala
- UPMC Hillman Cancer Center, Pittsburgh, PA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - Simion I Chiosea
- UPMC Hillman Cancer Center, Pittsburgh, PA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | | | - Julie E Bauman
- GW Cancer Center, The George Washington University School of Medicine and Health Sciences, Washington, DC
| |
Collapse
|
7
|
Achard V, Abdel-Aty H, Santana VD, Bebek M, Kroese TE, Orazem M, Dirix P, Ost P, Lancia A. Combining Radiotherapy with Immune Checkpoint Inhibitors for the Management of Muscle-Invasive Bladder Cancer: A Comprehensive Systematic Review from the Y-ECI ROSC EORTC group. Pract Radiat Oncol 2025:S1879-8500(25)00147-X. [PMID: 40412484 DOI: 10.1016/j.prro.2025.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/27/2025] [Accepted: 05/14/2025] [Indexed: 05/27/2025]
Abstract
OBJECTIVE This review aims to evaluate the efficacy and safety of combining immune checkpoint inhibitors (ICI) with RT in the treatment of non-metastatic MIBC, focusing on neoadjuvant and bladder preservation strategies. EVIDENCE ACQUISITION A systematic literature review was conducted from January 2000 to December 2023 using PubMed and Clinicaltrials.gov databases. Studies investigating outcomes after combining immunotherapy with RT for non-metastatic MIBC were selected. Data extraction included study identifiers, patient characteristics, intervention and control arm details, and primary outcomes. EVIDENCE SYNTHESIS Among 28 selected studies, diverse approaches were observed, including neoadjuvant and bladder preservation strategies. Neoadjuvant trials, such as RACE IT, explored the feasibility of combining ICI with RT before radical cystectomy, showing promising efficacy and safety outcomes. In bladder preservation strategies, completed as well as ongoing trials demonstrated acceptable toxicity profiles and promising early efficacy data for combining ICI with chemoradiotherapy or RT alone. CONCLUSION Combining ICI with RT holds significant promise as a treatment strategy for non-metastatic MIBC. Preliminary evidence suggests favorable efficacy and safety profiles, supporting further exploration and potential integration into standard care. Ongoing phase III trials will provide crucial insights into the comparative effectiveness of adding ICI to conventional definitive treatments. In the evolving landscape of MIBC management, the combination of ICI and RT has the potential to reshape therapeutic paradigms and improve outcomes for patients.
Collapse
Affiliation(s)
- Vérane Achard
- Department of Radiotherapy, Institut Bergonié, Bordeaux, France; Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| | - Hoda Abdel-Aty
- The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK; MRC Clinical Trials Unit, Institute of Clinical Trials & Methodology, University College London, London, UK
| | - Victor Duque Santana
- Department of Radiation Oncology, Quironsalud Madrid University Hospital, Spain; Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, 28223 Madrid, Spain
| | | | - Tiuri E Kroese
- Department of Radiation Oncology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Miha Orazem
- Institute of Oncology, Division of Radiation Oncology, Ljubljana, Slovenia and Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Piet Dirix
- Department of Radiation Oncology, Iridium Network, Wilrijk (Antwerp), Belgium
| | - Piet Ost
- Department of Radiation Oncology, Iridium Network, Wilrijk (Antwerp), Belgium
| | - Andrea Lancia
- Radiation Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
8
|
Liu S, He X, Liang S, Wu A, Liu L, Hu W. Carbon ion irradiation mobilizes antitumor immunity: from concept to the clinic. Radiat Oncol 2025; 20:85. [PMID: 40405246 PMCID: PMC12100795 DOI: 10.1186/s13014-025-02647-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 04/23/2025] [Indexed: 05/24/2025] Open
Abstract
Carbon ion radiotherapy (CIRT), a type of particle therapy, is at the forefront of clinical oncology treatments due to its superior physical properties and biological performance. Although CIRT has demonstrated outstanding therapeutic outcomes in clinical settings, the biological mechanisms underpinning its effects, particularly its immunogenic potential and the superiority of its induced antitumor immune response compared to photon radiotherapy, remain areas of active investigation. This review summarizes the latest research progress on the mechanisms of antitumor immune responses triggered by CIRT and discusses preclinical and clinical studies related to combined CIRT and immunotherapy (CCIT). Against the backdrop of extensive research and significant clinical efficacy achieved by combining radiotherapy with immunotherapy, this review provides a theoretical foundation for a better understanding of the superior tumor cell-killing effects of CIRT and the underlying immunological mechanisms. Further insights into the factors affecting the efficacy, toxic effects, and developmental limitations of this combination therapy mode will be instrumental in guiding the conduction of CCIT studies.
Collapse
Affiliation(s)
- Shanghai Liu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, China
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiangyang He
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, China
| | - Siqi Liang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, China
| | - Anqing Wu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, China.
| | - Lu Liu
- Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, China.
| | - Wentao Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
9
|
Noda Y, Atsumi N, Nakaya T, Iwai H, Tsuta K. High-Sensitivity PD-L1 Staining Using Clone 73-10 Antibody and Spatial Transcriptomics for Precise Expression Analysis in Non-Tumorous, Intraepithelial Neoplasia, and Squamous Cell Carcinoma of Head and Neck. Head Neck Pathol 2025; 19:65. [PMID: 40392349 DOI: 10.1007/s12105-025-01798-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Accepted: 04/24/2025] [Indexed: 05/22/2025]
Abstract
PURPOSE While immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 axis have improved outcomes in head and neck squamous cell carcinoma (HNSCC), eligibility criteria based on immunohistochemistry (IHC) target PD-1 solely. We aimed to evaluate the PD-L1 (CD274) expression using highly sensitive clone 73 - 10 and spatial transcriptomics (ST) analysis to elucidate the role of PD-L1 in HNSCC and thus potentially expand the pool of eligible patients. METHODS Immunohistochemical staining of 73 - 10, CD3, CD4, and CD8 were performed in 94 HNSCC clinical samples along with paired adjacent squamous intraepithelial neoplasm (SIN) and normal oral mucosa (NOM) samples. The 73 - 10 positivity was evaluated using a tumor cell score ≥ 1%, and the results were analyzed against clinicopathological features including CD4+ and CD8+ tumor-infiltrating lymphocytes (TILs), and clinical outcomes. Furthermore, ST and PD-L1 related pathway analysis was performed in 6 paired HNSCC, SIN and NOM samples. RESULTS The 73 - 10 detected-PD-L1 positivity was high in HNSCC (79%) compared to SIN (10%) and NOM (3%). 73 - 10+ correlated with high CD4+ TILs, as well as the independent prognostic factor of OS, DSS, and PFS of HNSCC (all p < 0.05). ST analysis revealed that the upregulated distribution of CD274 correlated with 73 - 10 positivity. Pathway analysis revealed a significant upregulation of CD274 and CD4 in HNSCC compared to SIN and NOM, and HIF-1α and IFN-γ as key regulators of PD-L1 expression in HNSCC. CONCLUSION Clone 73 - 10 is a relatively suitable candidate for identifying patients with PD-L1 expression eligible for ICI therapy. It demonstrates high sensitivity in detecting PD-L1 (CD274) in HNSCC, offering immunological and prognostic insights.
Collapse
Affiliation(s)
- Yuri Noda
- Department of Pathology and Laboratory Medicine, Kansai Medical University Hospital, 2-3-1 Shin-machi, Hirakata, Osaka, 573-1191, Japan.
- Department of Pathology, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan.
| | - Naho Atsumi
- Department of Pathology, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Takeo Nakaya
- Department of Pathology and Laboratory Medicine, Kansai Medical University Hospital, 2-3-1 Shin-machi, Hirakata, Osaka, 573-1191, Japan
- Department of Pathology, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Hiroshi Iwai
- Department of Otolaryngology, Head and Neck Surgery, Kansai Medical University Hospital, 2-3-1 Shinmachi, Hirakata, Osaka, 5731191, Japan
| | - Koji Tsuta
- Department of Pathology and Laboratory Medicine, Kansai Medical University Hospital, 2-3-1 Shin-machi, Hirakata, Osaka, 573-1191, Japan
- Department of Pathology, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| |
Collapse
|
10
|
Lin H, Ma J, Heng Y, Zhu X, Jin Q, Ding X, Tao L, Lu L. CD8 + T cells in patients with hypopharyngeal squamous cell carcinoma are susceptible to radiation-induced damage. Int J Cancer 2025; 156:2010-2023. [PMID: 39918311 DOI: 10.1002/ijc.35329] [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: 08/16/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 03/21/2025]
Abstract
Radiotherapy (RT) is a commonly used clinical management for hypopharyngeal squamous cell carcinoma (HPSCC), which represents the most unfavorable prognosis among all subtypes of head and neck squamous cell carcinoma. However, radiation may cause lymphopenia, a significantly adverse event with detrimental prognostic implications for patients. While CD8+ T cells are vital in tumor immunity, the specific effects of RT on CD8+ T cells as well as the underlying mechanisms have not been clearly elucidated. Here we found that subpopulations of peripheral T lymphocytes exhibited differential profiles in patients with HPSCC compared to healthy individuals both pre- and post-irradiation. Importantly, CD8+ T cells from HPSCC patients showed greater reduction of cytokine production, more severe proliferation defect, and increased apoptosis compared to those from healthy individuals after in vitro irradiation. Mechanistically, the ATM-Chk2 pathway mediated the enhanced apoptosis of CD8+ T lymphocytes from HPSCC patients upon irradiation. Therefore, our study demonstrated that CD8+ T cells in patients with HPSCC exhibit a higher susceptibility to radiation-induced damage compared to those in healthy individuals. The ATM-Chk2 pathway represents a potential immunotherapeutic target for safeguarding CD8+ T cells in HPSCC patients against radiation-induced apoptosis.
Collapse
Affiliation(s)
- Hanqing Lin
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Department of Otorhinolaryngology, Fujian Institute of Otorhinolaryngology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jingyu Ma
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Yu Heng
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Xiaoke Zhu
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Qiuyan Jin
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Xuping Ding
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Tao
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Liming Lu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Tseng I, Chen Y, Ai D, Zhu Z, Zhao W, Fan M, Li L, Zhu H, Li F, Xu Y, Yu L, Wang Z, Wang J, Liu Q, Deng J, Hao S, Fan Q, Ye J, Zhou J, Wu C, Tang H, Lin Q, Li J, Li Y, Wei S, Luo H, Cao J, Zheng X, Huang G, Zheng Y, Ping B, Zhao K. Radiation Dose-Volume Effects on Negative Tumor-Draining Lymph Nodes Affected T-cell Activation and Prognosis in Esophageal Cancer with Chemoradiotherapy. Clin Cancer Res 2025; 31:2024-2033. [PMID: 40053692 PMCID: PMC12079094 DOI: 10.1158/1078-0432.ccr-24-4123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/21/2025] [Accepted: 03/04/2025] [Indexed: 03/09/2025]
Abstract
PURPOSE A preclinical model found that elective nodal irradiation attenuated the efficacy of radiotherapy (RT) and radio-immunotherapy. However, limited clinical studies have explored the correlation between radiation dose-volume parameters of negative tumor-draining lymph nodes (TDLN) and T-cell activation/prognosis for patients with cancer treated with definitive radiochemotherapy. EXPERIMENTAL DESIGN Patients with locally advanced esophageal cancer undergoing definitive chemoradiotherapy (CRT) were selected from two prospective trials. Dose-volume parameters of TDLN as well as other lymphocyte-related organs at risk and lymphocyte subsets such as CD3-CD19+ B cells, CD8+CD28+ T cells, and activated T cells (CD3+CD8+HLA-DR+) before and at the end of RT were collected. Logistic analysis was utilized to correlate dose-volume parameters with reductions in lymphocyte subsets. Prognosis of TDLN irradiation was investigated through Kaplan-Meier analysis and Cox hazards models. RESULTS Among 512 patients, the median mean dose of TDLN and negative non-TDLN was 25.6 and 15.1 Gy, respectively. Multivariable analyses indicated that TDLN V15 >50% was an independent predictor of poorer local recurrence-free survival (HR, 1.31; P = 0.029) and distant metastasis-free survival (HR, 1.39; P < 0.001), as well as greater reductions in CD3-CD19+ B cells (OR, 1.98; P = 0.002), CD8+CD28+ T cells (OR, 3.42; P < 0.001), and CD3+CD8+HLA-DR+ T cells (OR, 4.67; P = 0.002) after RT. CONCLUSIONS A higher radiation dose-volume parameter of TDLNs in patients with esophageal cancer undergoing CRT was significantly associated with suppression of T-cell activation and a worse prognosis. Limiting the percentage of TDLN V15 may be beneficial for improving the prognosis of CRT with or without PD-1 inhibitors.
Collapse
Affiliation(s)
- Ihsuan Tseng
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Yun Chen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Dashan Ai
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Zhengfei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Weixin Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Min Fan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Ling Li
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Hongcheng Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Fangfang Li
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yang Xu
- Department of Medicine, Enhance Human Health Through Pharma Technology Innovation, Shanghai, China
| | - Lu Yu
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Zezhou Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Cancer Prevention, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Municipal Hospital Oncological Specialist Alliance, Shanghai, China
| | - Juanqi Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qi Liu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Jiaying Deng
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Shengnan Hao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Qingsong Fan
- Department of General Practice, Changhai Community Healthcare Center, Shanghai, China
| | - Jinjun Ye
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Nanjing, China
| | - Jialiang Zhou
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Chaoyang Wu
- Department of Radiation Oncology, Zhenjiang First People’s Hospital, Zhenjiang, China
| | - Huarong Tang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Qin Lin
- Department of Radiation Oncology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jiancheng Li
- Department of Radiation Oncology, Fujian Provincial Cancer Hospital, Fuzhou, China
| | - Yunhai Li
- Department of Radiation Oncology, Minhang Branch Hospital, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shihong Wei
- Department of Radiation Oncology, Gansu Cancer Hospital, Lanzhou, China
| | - Hui Luo
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang, China
| | - Jianzhong Cao
- Department of Radiation Oncology, Shanxi Cancer Hospital, Taiyuan, China
| | - Xiangpeng Zheng
- Department of Radiation Oncology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Guang Huang
- Department of Radiation Oncology, Hainan People’s Hospital, Haikou, China
| | - Yuwei Zheng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Bo Ping
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Kuaile Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| |
Collapse
|
12
|
Liang YL, Liu X, Shen LF, Hu GY, Zou GR, Zhang N, Chen CB, Chen XZ, Zhu XD, Yuan YW, Yang KY, Jin F, Hu WH, Xie FY, Huang Y, Han F, Tang LL, Mao YP, Lu LX, Sun R, He YX, Zhou YY, Long GX, Tang J, Chen LS, Zong JF, Jin T, Li L, Lin J, Huang J, Gong XY, Zhou GQ, Chen L, Li WF, Chen YP, Xu C, Lin L, Huang SH, Huang SW, Wang YQ, Huang CL, Feng HX, Hou M, Chen CH, Zheng SF, Li YQ, Hong SB, Jie YS, Li H, Yun JP, Zang SB, Liu SR, Lin QG, Li HJ, Tian L, Liu LZ, Zhao HY, Li JB, Lin AH, Liu N, Zhang Y, Guo R, Ma J, Sun Y. Adjuvant PD-1 Blockade With Camrelizumab for Nasopharyngeal Carcinoma: The DIPPER Randomized Clinical Trial. JAMA 2025; 333:1589-1598. [PMID: 40079940 PMCID: PMC11907361 DOI: 10.1001/jama.2025.1132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 01/23/2025] [Indexed: 03/15/2025]
Abstract
Importance Approximately 20% to 30% of patients with locoregionally advanced nasopharyngeal carcinoma (NPC) experience disease relapse despite definitive chemoradiotherapy. The programmed cell death 1 (PD-1) blockade camrelizumab has demonstrated considerable value in recurrent or metastatic NPC, while its role in locoregionally advanced NPC is unclear. Objective To evaluate the efficacy and safety of adjuvant camrelizumab for patients with locoregionally advanced NPC. Design, Setting, and Participants Randomized, open-label, multicenter, phase 3 clinical trial conducted from August 2018 to November 2021 at 11 centers in China and enrolling 450 patients with T4N1M0 or T1-4N2-3M0 NPC who had completed induction-concurrent chemoradiotherapy. The final date of follow-up was March 20, 2024. Interventions Patients were randomized (1:1) to receive adjuvant camrelizumab (200 mg intravenously once every 3 weeks for 12 cycles; n = 226) or observation (standard therapy group; n = 224). Main Outcomes and Measures The primary end point was event-free survival (freedom from distant metastasis, locoregional relapse, or death due to any cause). Secondary end points included distant metastasis-free survival, locoregional relapse-free survival, overall survival, safety, and health-related quality of life. Results Among the 450 participants (mean age, 45 [SD, 10] years; 24% women), after a median follow-up of 39 (IQR, 33-50) months, the camrelizumab group had a 3-year event-free survival rate of 86.9%, whereas the standard therapy group had a rate of 77.3% (stratified hazard ratio, 0.56; 95% CI, 0.36-0.89; P = .01). Grade 3 or 4 adverse events were reported in 23 patients (11.2%) in the camrelizumab and 7 (3.2%) in the standard therapy group. Reactive capillary endothelial proliferation was the most common adverse event related to camrelizumab, occurring in 85.8% of patients at grade 1 or 2, while 2% of patients had grade 3 or 4 events. There was no significant deterioration in quality of life associated with camrelizumab treatment. Conclusions and Relevance Adjuvant PD-1 blockade with camrelizumab significantly improved event-free survival with manageable toxicities, highlighting its potential role in the management of locoregionally advanced NPC. Trial Registration ClinicalTrials.gov Identifier: NCT03427827.
Collapse
Affiliation(s)
- Ye-Lin Liang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Xu Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
- Chinese Society of Clinical Oncology, Beijing, China
| | - Liang-Fang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Guang-Yuan Hu
- Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Guo-Rong Zou
- Department of Oncology, Panyu Central Hospital, Guangzhou, China
| | - Ning Zhang
- Department of Radiation Oncology, First People’s Hospital of Foshan, Foshan, China
| | - Chuan-Ben Chen
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Xiao-Zhong Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xiao-Dong Zhu
- Department of Radiation Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Ya-Wei Yuan
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Kun-Yu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Jin
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Wei-Han Hu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Fang-Yun Xie
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
- Chinese Society of Clinical Oncology, Beijing, China
| | - Ying Huang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Fei Han
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Ling-Long Tang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
- Chinese Society of Clinical Oncology, Beijing, China
| | - Yan-Ping Mao
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
- Chinese Society of Clinical Oncology, Beijing, China
| | - Li-Xia Lu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Rui Sun
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Yu-Xiang He
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Yang-Ying Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Guo-Xian Long
- Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Tang
- Department of Oncology, Panyu Central Hospital, Guangzhou, China
| | - Lu-Si Chen
- Department of Radiation Oncology, First People’s Hospital of Foshan, Foshan, China
| | - Jing-Feng Zong
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Ting Jin
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ling Li
- Department of Radiation Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Jie Lin
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Jing Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiu-Yun Gong
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Guan-Qun Zhou
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Lei Chen
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Wen-Fei Li
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Yu-Pei Chen
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
- Chinese Society of Clinical Oncology, Beijing, China
| | - Cheng Xu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Li Lin
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Shao-Hui Huang
- Department of Radiation Oncology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| | - Sai-Wei Huang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Ya-Qin Wang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Cheng-Long Huang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Hui-Xia Feng
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Min Hou
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Chun-Hua Chen
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Su-Fen Zheng
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Ying-Qing Li
- Emergency Department, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shu-Bin Hong
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu-Sheng Jie
- Department of Infectious Diseases, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hao Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing-Ping Yun
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Sheng-Bing Zang
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Song-Ran Liu
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qing-Guang Lin
- Department of Ultrasound, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hao-Jiang Li
- Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li Tian
- Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li-Zhi Liu
- Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hong-Yun Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ji-Bin Li
- Clinical Trials Center, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ai-Hua Lin
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Na Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
- Chinese Society of Clinical Oncology, Beijing, China
| | - Yuan Zhang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Rui Guo
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Jun Ma
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
- Chinese Society of Clinical Oncology, Beijing, China
| | - Ying Sun
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
- Chinese Society of Clinical Oncology, Beijing, China
| |
Collapse
|
13
|
Posner M. Curative Application of Immunotherapy in Untreated Head and Neck and Nasopharynx Cancer. JAMA 2025; 333:1584-1585. [PMID: 40080388 DOI: 10.1001/jama.2025.3012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Affiliation(s)
- Marshall Posner
- Tampa General Hospital Cancer Institute/Cancer Center of South Florida, Palm Springs
- University of South Florida/Morsani Medical College, Palm Springs
| |
Collapse
|
14
|
Haddad R, Fayette J, Teixeira M, Prabhash K, Mesia R, Kawecki A, Dechaphunkul A, Dinis J, Guo Y, Masuda M, Hsieh CY, Ghi MG, Vaz de Melo Sette C, Harrington K, Tahara M, Saba NF, Lau A, Jiang T, Yan Y, Ballinger M, Kaul M, Matheny C, Cuchelkar V, Wong DJ. Atezolizumab in High-Risk Locally Advanced Squamous Cell Carcinoma of the Head and Neck: A Randomized Clinical Trial. JAMA 2025; 333:1599-1607. [PMID: 40079944 PMCID: PMC11907359 DOI: 10.1001/jama.2025.1483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/30/2025] [Indexed: 03/15/2025]
Abstract
Importance Treating locally advanced squamous cell carcinoma of the head and neck (LA SCCHN) involves any combination of surgery, radiation, and chemotherapy, followed by routine monitoring for local recurrence or distant metastases. Given the poor patient outcomes, a significant unmet clinical need for improved treatment options remains. Objective To evaluate efficacy and safety of maintenance atezolizumab in patients with LA SCCHN at high risk of disease progression after multimodal definitive treatment. Design, Setting, and Participants IMvoke010 was a phase 3, global, double-blind, randomized clinical trial. Patients were recruited at 128 sites in 23 countries between April 3, 2018, and February 14, 2020 (clinical cutoff date: September 27, 2023). Eligible patients had LA SCCHN (stage IVa/IVb involving the oral cavity, larynx, hypopharynx, or human papillomavirus-negative oropharynx, or stage III human papillomavirus-positive oropharynx [AJCC Cancer Staging Manual, eighth edition]) without disease progression after multimodal definitive treatment. Intervention Patients were randomized (1:1) to receive atezolizumab 1200 mg or placebo every 3 weeks for 1 year or until disease recurrence, disease progression, unacceptable toxicity, or consent withdrawal. Main Outcomes and Measures The primary end point was investigator-assessed event-free survival. Other end points included overall survival and safety. Results Overall, 406 patients were randomized to receive atezolizumab (n = 203) or placebo (n = 203); baseline demographics were balanced between both treatment groups (<65 years, 142 [70.0%] vs 155 [76.4%]; male, 168 [82.8%] vs 174 [85.7%]; Asian, 68 [35.6%] vs 61 [31.0%]; Black, 1 [0.5%] vs 1 [0.5%]; and White, 121 [63.4%] vs 135 [68.5%], respectively). At clinical cutoff (median follow-up, 46.5 months), median investigator-assessed event-free survival was 59.5 months (95% CI, 46.8 to not estimable) with atezolizumab vs 52.7 months (95% CI, 41.4 to not estimable) with placebo (hazard ratio, 0.94; 95% CI, 0.70-1.26; P = .68). There was no difference in overall survival between atezolizumab and placebo (24-month overall survival, 82.0% vs 79.2%, respectively). No new or unexpected safety signals were identified. Conclusions and Relevance In this study, atezolizumab did not improve clinical outcomes in patients with LA SCCHN at high risk of disease progression after multimodal definitive treatment. These data contribute to evidence on the limited activity of checkpoint inhibitors in the global population of this disease setting. Overall, the role of immunotherapy for patients with LA SCCHN remains to be determined. Trial Registration ClinicalTrials.gov Identifier: NCT03452137.
Collapse
Affiliation(s)
| | - Jérôme Fayette
- Centre Leon Berard, Département d’Hématologie et d’Oncologie, Lyon, France
| | - Maria Teixeira
- IPO de Coimbra, Servico de Oncologia Medica, Coimbra, Portugal
| | - Kumar Prabhash
- Tata Memorial Hospital, Department of Medical Oncology, Mumbai, India
| | - Ricard Mesia
- Catalan Institute of Oncology, B-ARGO Group, IGTP, Badalona, Catalonia, Spain
| | | | - Arunee Dechaphunkul
- Unit of Medical Oncology, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - José Dinis
- IPO do Porto, Servico de Oncologia Medica, Porto, Portugal
| | - Ye Guo
- Shanghai East Hospital, Tongji University, Shanghai, China
| | - Muneyuki Masuda
- National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Ching-Yun Hsieh
- China Medical University Hospital, Oncology and Hematology Office Critical Care Center, Taichung, Taiwan
| | | | | | - Kevin Harrington
- The Royal Marsden Hospital/The Institute of Cancer Research, London, United Kingdom
| | - Makoto Tahara
- National Cancer Center Hospital East, Kashiwa, Japan
| | - Nabil F. Saba
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Agnes Lau
- Genentech, Inc, South San Francisco, California
| | - Tao Jiang
- Genentech, Inc, South San Francisco, California
| | - Yibing Yan
- Genentech, Inc, South San Francisco, California
| | | | - Monika Kaul
- Genentech, Inc, South San Francisco, California
| | | | | | - Deborah J. Wong
- Division of Hematology-Oncology, University of California Los Angeles (UCLA)
| |
Collapse
|
15
|
Xu C, Zhou GQ, Li WF, Hu DS, Chen XZ, Lin SJ, Jin F, Huang XQ, Peng G, Huang J, Wu Y, Tao CJ, Li JB, Lin AH, Zhao HY, Hong SB, Huang HL, Tang LL, Peng YL, Shi KF, Chen L, Qi LP, Yang KY, Shen LF, Sun Y, Ma J. Nivolumab combined with induction chemotherapy and radiotherapy in nasopharyngeal carcinoma: A multicenter phase 2 PLATINUM trial. Cancer Cell 2025; 43:925-936.e4. [PMID: 40020668 DOI: 10.1016/j.ccell.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/13/2024] [Accepted: 01/31/2025] [Indexed: 03/03/2025]
Abstract
Severe toxicities caused by concurrent cisplatin are a critical problem in nasopharyngeal carcinoma (NPC) treatment. In this phase 2 multicenter PLATINUM trial (NCT03984357), we recruited 152 NPC patients who received 12-cycle nivolumab plus induction chemotherapy and radiotherapy without concurrent cisplatin. After a median follow-up of 43 months, the 3-year failure-free survival (FFS) was 88.5% (95% confidence interval [CI], 83.4%-93.8%) and the 3-year overall survival was 97.9%. An early clearance of Epstein-Barr virus (EBV) DNA after induction-phase treatment was associated with FFS benefit. Sixty (40.2%) and eight (5.2%) patients had acute and late grade 3-4 adverse events (AEs), respectively. Most patients had good tolerance to AE-associated frequency (68.0%-96.7%), severity (56.0%-98.6%), and interference (58.0%-98.0%); 86.7%-100.0% of quality-of-life domains showed either no clinically meaningful deterioration or a rapid recovery. Nivolumab plus induction chemotherapy and radiotherapy demonstrated efficacious anti-tumor activity, low toxicity, and favorable tolerability and quality-of-life for NPC patients.
Collapse
Affiliation(s)
- Cheng Xu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510000, P.R. China
| | - Guan-Qun Zhou
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510000, P.R. China
| | - Wen-Fei Li
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510000, P.R. China
| | - De-Sheng Hu
- Department of Radiotherapy, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, P.R. China
| | - Xiao-Zhong Chen
- Department of Head and Neck Tumor Radiotherapy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, P.R. China
| | - Shao-Jun Lin
- Department of Radiation Oncology, Cancer Hospital of Fujian Medical University (Fujian Provincial Cancer Hospital), Fuzhou, Fujian 350014, P.R. China
| | - Feng Jin
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Xin-Qiong Huang
- Department of Radiation Oncology, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Gang Peng
- Department of Oncology, Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jing Huang
- Department of Oncology, Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yuan Wu
- Department of Radiotherapy, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, P.R. China
| | - Chang-Juan Tao
- Department of Head and Neck Tumor Radiotherapy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, P.R. China
| | - Ji-Bin Li
- Clinical Trials Center, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510000, P.R. China
| | - Ai-Hua Lin
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Hong-Yun Zhao
- Department of Medical Oncology, and Department of Clinical Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510000, P.R. China
| | - Shu-Bin Hong
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Hui-Ling Huang
- Department of Cardiology, Cardiac Prevention and Assessment Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Ling-Long Tang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510000, P.R. China
| | - Ying-Lin Peng
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510000, P.R. China
| | - Ke-Fu Shi
- Nursing Division, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510000, P.R. China
| | - Liu Chen
- Nursing Division, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510000, P.R. China
| | - Li-Ping Qi
- Nursing Division, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510000, P.R. China
| | - Kun-Yu Yang
- Department of Oncology, Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China.
| | - Liang-Fang Shen
- Department of Radiation Oncology, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China.
| | - Ying Sun
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510000, P.R. China; Chinese Society of Clinical Oncology, Beijing 100000, P.R. China.
| | - Jun Ma
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510000, P.R. China; Chinese Society of Clinical Oncology, Beijing 100000, P.R. China.
| |
Collapse
|
16
|
Hut AR, Boia ER, Para D, Iovanescu G, Horhat D, Mikša L, Chiriac M, Galant R, Motofelea AC, Balica NC. Laryngeal Cancer in the Modern Era: Evolving Trends in Diagnosis, Treatment, and Survival Outcomes. J Clin Med 2025; 14:3367. [PMID: 40429363 PMCID: PMC12112285 DOI: 10.3390/jcm14103367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2025] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Laryngeal cancer (LC), predominantly squamous cell carcinoma (SCC), represents a considerable health burden worldwide. Tumour subsite heterogeneity (supraglottic, glottic, subglottic) influences clinical behavior and outcomes. This review synthesizes current knowledge on epidemiology, risk factors, diagnostics, histological variants, biomarkers, treatment modalities, and survival. Results: This narrative review synthesizes current literature on the epidemiology, risk factors, diagnosis, histological variants, biomarkers, and prognosis of LC. The review highlights the critical influence of tumour sites (supraglottic, glottic, subglottic) on metastatic patterns and survival. Key risk factors of LC include tobacco and alcohol use, human papillomavirus (HPV) infection, and occupational exposures. The diagnostic process encompasses clinical examination, endoscopy, biopsy, and imaging. Several biomarkers that aid in diagnosis, treatment plan determination, and prognosis prediction have been established. These biomarkers include long noncoding RNAs, cell cycle regulators, apoptosis regulators, oncogenes, tumour suppressor genes, growth factor pathway components, angiogenic factors, structural proteins, sex hormone receptors, and immunological markers. Current treatment modalities range from organ-preserving surgery and radiotherapy to combined chemoradiotherapy and total laryngectomy. Finally, survival data are presented and stratified by stage and subsite. Conclusions: The review underscores the need for a multidisciplinary approach to LC management, integrating clinical, pathological, and molecular information to optimize patient outcomes.
Collapse
Affiliation(s)
- Alexandru-Romulus Hut
- Department of Doctoral Studies, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (A.-R.H.); (D.P.); (A.C.M.)
- ENT Department, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (G.I.); (D.H.); (L.M.); (M.C.); (N.C.B.)
| | - Eugen Radu Boia
- ENT Department, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (G.I.); (D.H.); (L.M.); (M.C.); (N.C.B.)
- ENT Department, Emergency City Hospital, 300254 Timisoara, Romania
| | - Diana Para
- Department of Doctoral Studies, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (A.-R.H.); (D.P.); (A.C.M.)
| | - Gheorghe Iovanescu
- ENT Department, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (G.I.); (D.H.); (L.M.); (M.C.); (N.C.B.)
| | - Delia Horhat
- ENT Department, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (G.I.); (D.H.); (L.M.); (M.C.); (N.C.B.)
- ENT Department, Emergency City Hospital, 300254 Timisoara, Romania
| | - Loredan Mikša
- ENT Department, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (G.I.); (D.H.); (L.M.); (M.C.); (N.C.B.)
| | - Maria Chiriac
- ENT Department, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (G.I.); (D.H.); (L.M.); (M.C.); (N.C.B.)
| | - Raphaël Galant
- Hôpital Européen Georges-Pompidou, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, 20 Rue Leblanc, 75015 Paris, France;
| | - Alexandru Catalin Motofelea
- Department of Doctoral Studies, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (A.-R.H.); (D.P.); (A.C.M.)
- Center for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Nicolae Constantin Balica
- ENT Department, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (G.I.); (D.H.); (L.M.); (M.C.); (N.C.B.)
- ENT Department, Emergency City Hospital, 300254 Timisoara, Romania
| |
Collapse
|
17
|
Goss G, Ciuleanu T, Ramlau R, Renouf DJ, Chu Q, Kalinka E, Sawrycki P, Bramson J, Nelson BH, Crabbé R, LaCasse E, Lo B, Sahlender DA, Crompton P, Brichory F, Piggott L, Schenker M, Juergens R. Xevinapant plus avelumab in advanced solid tumours, with a dose expansion in advanced non-small-cell lung cancer: exploratory biomarker, safety and efficacy analyses from an open-label, nonrandomised phase Ib study. Ther Adv Med Oncol 2025; 17:17588359251332154. [PMID: 40351326 PMCID: PMC12062605 DOI: 10.1177/17588359251332154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/18/2025] [Indexed: 05/14/2025] Open
Abstract
Background Xevinapant, an inhibitor of apoptosis protein (IAP) inhibitor, has shown promising activity in combination with anticancer agents, including radiotherapy, and, in preclinical studies, anti-PD-(L)1 antibodies. This, in part, is due to its ability to restore apoptosis and increase antitumour immunity. Objectives We report efficacy, safety and exploratory biomarker analyses of xevinapant plus avelumab (anti-PD-L1) in a two-part, open-label, nonrandomised, phase Ib study. Design Part A assessed patients with advanced solid tumours who received xevinapant (100, 150, 200 or 250 mg/day, with no random allocation, on Days 1-10 and 15-24) in combination with avelumab (10 mg/kg) on Days 1 and 15 in 28-day cycle. Part B assessed patients with advanced non-small-cell lung cancer (NSCLC) who received xevinapant at the recommended phase II dose (RP2D) plus avelumab (maximum 26 cycles). Methods Part A assessed the safety and tolerability of the combination and established the maximum tolerated dose (MTD) and RP2D of xevinapant. Part B assessed the antitumour activity of xevinapant at the RP2D combined with avelumab compared with a historical control (avelumab alone). Exploratory biomarker analyses were also conducted. Results In part A (n = 16), xevinapant 200 mg/day was established as the RP2D with avelumab and the MTD was not reached. The most common treatment-emergent adverse events (TEAEs) irrespective of xevinapant dose were nausea and fatigue (n = 11 (68.8%) each). In part B (n = 38; four patients received prior anti-PD-(L)1 antibody), the objective response rate (ORR) was 10.5% (95% confidence interval (CI), 2.9-24.8; partial response, n = 4) and the most common TEAE was decreased appetite (n = 13 (34.2%)). Levels of plasma IL-10, IL-1β, IL-13 and CD8+ T cells increased during the study, and circulating levels of CD4+ T cells and Tregs increased during cycle 1. Macrophage-related gene expression signatures increased in patients with a partial response or stable disease. Low baseline Ki-67 expression in tumour samples correlated with a partial response. Conclusion The RP2D of xevinapant with avelumab was established; however, the ORR was not superior to the historical control (avelumab alone). The combination had a manageable safety profile in both study parts. Biomarker analyses provide insights into drivers associated with efficacy in patients with NSCLC receiving xevinapant plus avelumab. Trial registration NCT03270176 (https://clinicaltrials.gov/study/NCT03270176). Registered on ClinicalTrials.gov on 29 August 2017.
Collapse
Affiliation(s)
- Glenwood Goss
- Department of Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H8L6, Canada
| | - Tudor Ciuleanu
- Department of Oncology, ‘Iuliu Haţieganu’ University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Rodryg Ramlau
- Oncology Department, Poznan University of Medical Sciences, Poznan, Poland
| | - Daniel J. Renouf
- Department of Medicine, Faculty of Medicine, University of British Columbia, BC Cancer, Vancouver, BC, Canada
| | - Quincy Chu
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB, Canada
| | - Ewa Kalinka
- Department of Oncology, Polish Mother’s Memorial Hospital, Lodz, Poland
| | - Piotr Sawrycki
- Wojewódzki Szpital Zespolony im. L. Rydygiera w Toruniu, Torun, Poland
| | - Jonathan Bramson
- Centre for Discovery in Cancer Research and Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Brad H. Nelson
- Deeley Research Centre, BC Cancer – Victoria, Victoria, BC, Canada
| | | | | | - Bryan Lo
- Division of Anatomical Pathology, The Ottawa Hospital, Ottawa, ON, Canada
| | | | | | | | - Luke Piggott
- Debiopharm International SA, Lausanne, Switzerland
| | - Michael Schenker
- Oncology Center Sf Nectarie, Craiova, Romania
- Medical Oncology, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Rosalyn Juergens
- Department of Oncology, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
18
|
Zahnreich S, Bhatti A, Ahmad B, Drabke S, Kaufmann J, Schmidberger H. Effects of Cisplatin on the Radiation Response and DNA Damage Markers in Peripheral Blood Lymphocytes Ex Vivo. Cells 2025; 14:682. [PMID: 40422185 DOI: 10.3390/cells14100682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/30/2025] [Accepted: 05/07/2025] [Indexed: 05/28/2025] Open
Abstract
Platinum-based radiochemotherapy is associated with hematologic side effects, impacting patient outcomes. However, the clinical mechanisms of cisplatin and its interaction with ionizing radiation (IR), including in biodosimetry for radiotherapy, have not yet been fully clarified. For this purpose, healthy donors' peripheral blood lymphocytes (PBLs) were pretreated with cisplatin in a pulse (1-4 h) or continuous (24 h) regimen followed by X-rays. DNA damage was assessed as DNA double-strand breaks using repair foci of γH2AX and 53BP1 after 0.5 h and 24 h in G1 PBLs and a proliferation-based cytokinesis-block micronucleus assay. Additionally, cell death and proliferation activity were measured. Unlike a 1 h pulse, a 24 h cisplatin pretreatment caused a concentration-dependent increase in cisplatin-induced foci while decreasing IR-induced foci, especially 24 h after irradiation. This was accompanied by increased apoptosis, with cisplatin and IR having additive effects. Both genotoxins alone caused a dose-dependent increase in micronuclei, while cisplatin significantly reduced binuclear cells, especially after the 24 h treatment, leading to lower micronuclei frequencies post-irradiation. Our results show that prolonged cisplatin exposure, even at low concentrations, impacts the vitality and division activity of PBLs, with significantly stronger effects post-irradiation. This has major implications and must be considered for the detection of DNA damage-associated biomarkers in PBLs used in clinical prediction or biodosimetry during radiotherapy.
Collapse
Affiliation(s)
- Sebastian Zahnreich
- Department of Radiation Oncology and Radiation Therapy, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Aisha Bhatti
- Department of Radiation Oncology and Radiation Therapy, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Barea Ahmad
- Department of Radiation Oncology and Radiation Therapy, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Sophia Drabke
- Department of Radiation Oncology and Radiation Therapy, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Justus Kaufmann
- Department of Radiation Oncology and Radiation Therapy, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Heinz Schmidberger
- Department of Radiation Oncology and Radiation Therapy, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| |
Collapse
|
19
|
Rosenberg AJ, Juloori A, Jelinek MJ, Agrawal N, Cursio JF, Cipriani N, Lingen MW, Izumchenko E, Katipally R, Chin J, Ginat D, Pasternak-Wise O, Gooi Z, Blair E, Pearson AT, Haraf DJ, Vokes EE. Neoadjuvant Nivolumab Plus Chemotherapy Followed by Response-Stratified Chemoradiation Therapy in HPV-Negative Head and Neck Cancer: The DEPEND Phase 2 Nonrandomized Clinical Trial. JAMA Oncol 2025; 11:492-501. [PMID: 40048190 PMCID: PMC11886870 DOI: 10.1001/jamaoncol.2025.0081] [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: 11/11/2024] [Accepted: 01/09/2025] [Indexed: 03/09/2025]
Abstract
Importance Neoadjuvant immunotherapy in human papillomavirus (HPV)-negative locoregionally advanced (LA) head and neck squamous cell carcinoma (HNSCC) appears promising, yet its role in nonsurgical treatment for head and neck cancer remains undefined. Neoadjuvant nivolumab plus chemotherapy followed by response-stratified de-escalated chemoradiation therapy (CRT) in HPV-negative LA stage IVa/b HNSCC may improve treatment efficacy while reducing treatment-related toxic effects. Objective To determine the deep response rate and tolerability of neoadjuvant nivolumab plus chemotherapy followed by response-stratified CRT in nonvirally mediated stage IVa/b HNSCC. Design, Setting, and Participants In this investigator-initiated phase 2 nonrandomized clinical trial conducted at a single academic center, patients with stage IVa/b (American Joint Committee on Cancer Tumor Classification, 8th edition) HPV-negative LA HNSCC were enrolled between 2019 and 2022. Data were analyzed from February 2023 to January 2024. Interventions The DEPEND trial evaluated neoadjuvant nivolumab plus carboplatin and paclitaxel, followed by response-stratified CRT. Patients with 50% or greater reduction per Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 received de-escalated CRT to 66 Gy with elimination of elective nodal volumes; patients with less than 50% reduction received standard CRT to 70 to 75 Gy. Adjuvant nivolumab was administered for 9 cycles. Main Outcomes and Measures The primary end point was deep response rate (DRR; 50% or greater shrinkage per RECIST version 1.1) following neoadjuvant nivolumab plus chemotherapy. Secondary end points included progression-free survival (PFS), overall survival (OS), locoregional control, and distant control. Exploratory end points included acute toxic effects in patients who received response-adapted de-escalated CRT. Results Of 36 included patients, 28 (78%) were male, and the median (range) age was 58.9 (27-77) years. All patients started treatment and were available for analysis. The median (range) follow-up was 20 (13-40) months. The primary end point was met, with a DRR following neoadjuvant nivolumab/chemotherapy of 53% (95% CI, 35-70). The objective response rate was 86% (95% CI, 71-95). A total of 19 received de-escalated CRT and 16 received standard CRT. PFS and OS at 2 years were 66% (95% CI, 34-76) and 73% (95% CI, 52-86), respectively. The most common treatment-emergent adverse events for de-escalated and standard CRT were mucositis (14 of 19 [74%] and 15 of 16 [94%], respectively), radiation dermatitis (13 of 19 [68%] and 14 of 16 [88%], respectively), and dry mouth (7 of 19 [37%] and 10 of 16 [63%], respectively). Conclusions and Relevance In this phase 2 nonrandomized clinical trial, neoadjuvant nivolumab/chemotherapy led to deep responses in 53% of patients with HPV-negative LA stage IVa/b HNSCC, and response-adapted de-escalated CRT led to favorable survival with lower acute toxic effects among deep responders. Trial Registration ClinicalTrials.gov Identifier: NCT03944915.
Collapse
Affiliation(s)
- Ari J. Rosenberg
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, Illinois
- University of Chicago Comprehensive Cancer Center, Chicago, Illinois
| | - Aditya Juloori
- University of Chicago Comprehensive Cancer Center, Chicago, Illinois
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | | | - Nishant Agrawal
- University of Chicago Comprehensive Cancer Center, Chicago, Illinois
- Section of Otolaryngology-Head and Neck Surgery, University of Chicago, Chicago, Illinois
| | - John F. Cursio
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois
| | - Nicole Cipriani
- University of Chicago Comprehensive Cancer Center, Chicago, Illinois
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Mark W. Lingen
- University of Chicago Comprehensive Cancer Center, Chicago, Illinois
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Evgeny Izumchenko
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, Illinois
- University of Chicago Comprehensive Cancer Center, Chicago, Illinois
| | - Rohan Katipally
- University of Chicago Comprehensive Cancer Center, Chicago, Illinois
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Jeffrey Chin
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, Illinois
| | - Daniel Ginat
- University of Chicago Comprehensive Cancer Center, Chicago, Illinois
- Department of Radiology, University of Chicago, Chicago, Illinois
| | - Olga Pasternak-Wise
- University of Chicago Comprehensive Cancer Center, Chicago, Illinois
- Department of Radiology, University of Chicago, Chicago, Illinois
| | - Zhen Gooi
- Section of Otolaryngology-Head and Neck Surgery, University of Chicago, Chicago, Illinois
| | - Elizabeth Blair
- University of Chicago Comprehensive Cancer Center, Chicago, Illinois
- Section of Otolaryngology-Head and Neck Surgery, University of Chicago, Chicago, Illinois
| | - Alexander T. Pearson
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, Illinois
- University of Chicago Comprehensive Cancer Center, Chicago, Illinois
| | - Daniel J. Haraf
- University of Chicago Comprehensive Cancer Center, Chicago, Illinois
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Everett E. Vokes
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, Illinois
- University of Chicago Comprehensive Cancer Center, Chicago, Illinois
| |
Collapse
|
20
|
Yang J, Liu C, Zuo Z, Cao F, Zhang Z, Wu B, Qin Y, Wen L, Wei J, Xiao G, Xing S, Qu Y, Huang L, Wang X, Wang B, Yang K, Jiang K. Neoadjuvant chemoradiotherapy plus sequential tislelizumab followed by surgery for esophageal carcinoma (CRISEC study): A single-arm, bicentric, phase 2 trial. Radiother Oncol 2025; 206:110797. [PMID: 39978682 DOI: 10.1016/j.radonc.2025.110797] [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: 10/16/2024] [Revised: 02/15/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND AND PURPOSE To explore the efficacy and safety of neoadjuvant chemoradiotherapy (NCRT) plus sequential tislelizumab followed by surgery for esophageal squamous cell carcinoma (ESCC). MATERIALS AND METHODS This single-arm, bicentric, phase 2 trial enrolled patients with resectable or potentially resectable thoracic ESCC to receive neoadjuvant radiotherapy (41.4 Gy in 23 fractions) with concurrent chemotherapy (albumin-bound paclitaxel, 50-100 mg/m2, and carboplatin, area under the curve of 2 mg/ml/min, once weekly, five times) plus sequential tislelizumab (200 mg Q3W, three cycles) followed by surgery. The primary endpoint was pathologic complete response (pCR) rate. The secondary endpoints included safety, R0 resection rate, major pathologic response (MPR) rate, disease-free survival (DFS), and overall survival (OS). RESULTS Of the 30 patients enrolled from January 2021 to October 2022, 24 (80.0 %) completed planned surgery and gained R0 resection (100 %). Among the 24 patients, nine (37.5 %) achieved pCR and 21 (87.5 %) achieved MPR. Ten patients (35.7 %) developed grade 3-4 toxicities during tislelizumab therapy, including lymphopenia (32.1 %), neutropenia (3.6 %), and thrombocytopenia (3.6 %). Grade 5 hematemesis occurred in two patients and both were attributed to aortic invasion. Three patients (12.5 %) developed grade 3 postoperative complications, including pulmonary infection (8.3 %) and hoarseness (4.2 %). After a median follow-up of 35.4 months, the 2-year OS and DFS rates were 83.3 % and 79.2 %, respectively. CONCLUSION Sequential tislelizumab after NCRT in ESCC is safe and feasible. Further study is warranted to validate the efficacy of this combination mode.
Collapse
Affiliation(s)
- Jinsong Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Cui Liu
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhigang Zuo
- Department of Oncology, Shiyan People's Hospital, Hubei University of Medicine, Shiyan, China
| | - Fengjun Cao
- Department of Oncology, Shiyan People's Hospital, Hubei University of Medicine, Shiyan, China
| | - Zhanjie Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Bian Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - You Qin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Lu Wen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Jielin Wei
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Guangqin Xiao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Shijie Xing
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Qu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Huang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaolin Wang
- Department of Thoracic Surgery, Subei People's Hospital, Yangzhou University, Yangzhou, China
| | - Buhai Wang
- Department of Oncology, Subei People's Hospital, Yangzhou University, Yangzhou, China
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China.
| | - Ke Jiang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
21
|
Sim ES, Nguyen HCB, Hanna GJ, Uppaluri R. Current Progress and Future Directions of Immunotherapy in Head and Neck Squamous Cell Carcinoma: A Narrative Review. JAMA Otolaryngol Head Neck Surg 2025; 151:521-528. [PMID: 40048196 DOI: 10.1001/jamaoto.2024.5254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Importance For decades, the 3 therapeutic pillars for head and neck squamous cell carcinoma (HNSCC) have been radiation therapy, chemotherapy, and surgery. In recent years, a fourth pillar, immunotherapy, has shifted the existing paradigm of oncologic care by improving survival outcomes. This narrative review highlights key completed and ongoing clinical trials that have led to new therapeutic approaches and are aiming to further alter the current standard of care. Observations Immunotherapy in HNSCC first saw success in phase 3 clinical trials with immune checkpoint inhibitors (ICIs) for programmed cell death 1 protein in patients with recurrent or metastatic (R/M) disease. However, only approximately 15% to 20% of patients with R/M HNSCC achieve durable responses. Subsequent trials aimed to broaden ICIs to the definitive or curative setting, in combination with established chemoradiation modalities. These studies have yielded disappointing results, raising concerns that concurrent administration of ICI with chemoradiation- or radiation-induced attenuation of immune responses may contribute to lack of efficacy. Therefore, recent studies have attempted to introduce ICI sequentially, either prior to standard of care surgery in the neoadjuvant setting or following definitive treatment in the adjuvant or maintenance setting. These trials have demonstrated mixed results but with promising initial results from early phase neoadjuvant trials demonstrating early signals of response. Further trials are currently underway with various combinatorial approaches in the neoadjuvant and adjuvant settings to assess response rates and survival. Conclusions and Relevance The introduction of ICIs has brought a dramatic shift in the treatment landscape of HNSCC. Completed trials have provided new hope for patients, but failures in several settings suggest that further studies based on a biologic understanding of immune responses are required to expand immunotherapeutic approaches.
Collapse
Affiliation(s)
- Edward S Sim
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts
- Division of Otolaryngology-Head and Neck Surgery, Brigham and Women's Hospital, Boston, Massachusetts
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Hoang C B Nguyen
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts
- Division of Otolaryngology-Head and Neck Surgery, Brigham and Women's Hospital, Boston, Massachusetts
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Glenn J Hanna
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ravindra Uppaluri
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts
- Division of Otolaryngology-Head and Neck Surgery, Brigham and Women's Hospital, Boston, Massachusetts
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
22
|
Hausmann P, Zschaeck S, Furth C, Nikulin P, Cegla P, Roohani S, Lombardo E, Kazmierska J, Albert NL, Holzgreve A, Strouthos I, Belka C, Landry G, Cholewinski W, Kotzerke J, Baumann M, Krause M, Zips D, van den Hoff J, Hofheinz F. Tumor Asphericity in FDG PET Is an Independent Prognostic Parameter Improving Risk Stratification in Patients with Head and Neck Squamous Cell Carcinoma. J Nucl Med 2025; 66:686-691. [PMID: 40081960 PMCID: PMC12051765 DOI: 10.2967/jnumed.124.268972] [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: 10/27/2024] [Accepted: 02/07/2025] [Indexed: 03/16/2025] Open
Abstract
Tumor asphericity in 18F-FDG PET is a prognostic marker that has been investigated in small pilot studies of patients with head and neck squamous cell carcinoma (HNSCC). Here, we investigated the prognostic role of asphericity in a large multicenter database of patients with HNSCC treated with primary radiotherapy or chemoradiation and assessed its independent prognostic value. Methods: In total, 1,104 patients were included in this analysis. All received pretreatment 18F-FDG PET scans. Clinical risk factors were evaluated, and quantitative PET parameters SUVmax, metabolic tumor volume (MTV), total lesion glycolysis, and asphericity were calculated. Primary study endpoints were overall survival (OS) and locoregional control (LRC). Uni- and multivariate Cox regression analyses were performed. Additionally, asphericity was combined with the best-established quantitative PET parameter of MTV, and the combinatory approach of using asphericity and MTV was compared with the use of only asphericity or MTV by bootstrap analyses. Results: Asphericity showed only a modest correlation with the established PET parameters of MTV, SUVmax, and total lesion glycolysis. On univariate testing asphericity was strongly associated with the outcome of patients (LRC and OS with P < 0.001). In multivariate testing of all imaging parameters that were not highly correlated, both MTV and asphericity showed a significant association with LRC (P < 0.001 for MTV and P = 0.021 for asphericity) and OS (P < 0.001 for MTV and asphericity). Asphericity and MTV were binarized and combined for risk stratification, and the prognostic value of the combination was compared with the prognostic value of individual parameters. Bootstrapping revealed significantly better performance by the combinatory approach when compared with MTV (P = 0.012 for LRC and P < 0.001 for OS) and asphericity with regard to OS (P < 0.001) but not for LRC (P = 0.53). Conclusion: We were able to show that asphericity bears independent prognostic value and significantly improves risk stratification when combined with MTV in a comprehensive retrospective cohort of HNSCC patients.
Collapse
Affiliation(s)
- Patrick Hausmann
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany;
| | - Sebastian Zschaeck
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- BIH Charité (Junior) Clinician Scientist Program, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, Berlin, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Cancer Consortium, partner site Dresden, and German Cancer Research Center, Heidelberg, Germany
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Christian Furth
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Pavel Nikulin
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Paulina Cegla
- Department of Nuclear Medicine, Greater Poland Cancer Centre, Poznan, Poland
| | - Siyer Roohani
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- BIH Charité (Junior) Clinician Scientist Program, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, Berlin, Germany
- German Cancer Consortium, partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Elia Lombardo
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Joanna Kazmierska
- Electroradiology Department, University of Medical Sciences, Poznan, Poland
- Radiotherapy Department II, Greater Poland Cancer Centre, Poznan, Poland
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Iosif Strouthos
- Department of Radiation Oncology, German Oncology Center, European University Cyprus, Limassol, Cyprus
| | - Claus Belka
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center, partner site Munich, Munich, Germany
- German Cancer Consortium, partner site Munich, Munich, Germany
| | - Guillaume Landry
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center, partner site Munich, Munich, Germany
- German Cancer Consortium, partner site Munich, a partnership between DKFZ and LMU University Hospital Munich, Munich, Germany
| | - Witold Cholewinski
- Department of Nuclear Medicine, Greater Poland Cancer Centre, Poznan, Poland
- Electroradiology Department, University of Medical Sciences, Poznan, Poland
| | - Jorg Kotzerke
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Michael Baumann
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Division of Radiooncology/Radiobiology, German Cancer Research Center, Heidelberg, Germany
| | - Mechthild Krause
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Cancer Consortium, partner site Dresden, and German Cancer Research Center, Heidelberg, Germany
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- National Center for Tumor Diseases, partner site Dresden, Dresden, Germany, German Cancer Research Center, Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology, Dresden, Germany; and
| | - Daniel Zips
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- German Cancer Consortium, partner site Dresden, and German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, Berlin, Germany
- National Tumor Center Berlin, partner site Berlin, Berlin, Germany, German Cancer Research Center, Heidelberg, Germany, Charité Comprehensive Cancer, Charité-Universitätsmedizin Berlin, Berlin, Germany, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany, and Max-Delbrück-Centrum für Molekulare Medizin, Helmholtz Association, Berlin, Germany
| | - Jörg van den Hoff
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Bavarian Cancer Research Center, partner site Munich, Munich, Germany
- German Cancer Consortium, partner site Munich, Munich, Germany
| | - Frank Hofheinz
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| |
Collapse
|
23
|
Chen L, He JN, Zhao SJ, Peng LP, Mo DC, Yin SH. Efficacy and safety of PD-1/PD-L1 inhibitors combined with standard of care for locally advanced head and neck squamous cell carcinoma: A meta-analysis of randomized controlled trials. Crit Rev Oncol Hematol 2025; 209:104668. [PMID: 39978426 DOI: 10.1016/j.critrevonc.2025.104668] [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: 01/13/2025] [Revised: 02/05/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025] Open
Abstract
OBJECTIVES Chemoradiotherapy (CRT) or radiotherapy (RT) combined with cetuximab (for cisplatin-ineligible patients) is the standard of care (SoC) for locally advanced head and neck squamous cell carcinoma (LA-HNSCC). This study investigates whether adding programmed cell death-1/programmed cell death-ligand 1 (PD-1/PD-L1) immune checkpoint inhibitors (ICIs) to standard therapy improves survival in patients with LA-HNSCC. METHODS A comprehensive search of PubMed, Embase, and the Cochrane Library identified randomized controlled trials (RCTs) evaluating PD-1/PD-L1 inhibitors plus SoC compared with SoC alone for LA-HNSCC. The primary endpoints were progression-free survival (PFS), overall survival (OS), locoregional event-free survival (LEFS), and distant metastasis-free survival (DMFS) at the 1-year and 2-year time points, as well as the incidence of grade 3 or higher adverse events (AEs). RESULTS Four RCTs encompassing 1818 patients met the inclusion criteria. Compared with SoC alone, PD-1/PD-L1 inhibitors combined with SoC did not significantly improve 1-year or 2-year PFS, OS, LEFS, or DMFS (all p > 0.05). Subgroup analyses further showed no survival benefit at 2 years in the ICI + CRT, ICI + RT-cetuximab, anti-PD-1, or anti-PD-L1 subgroups. Additionally, there was no statistically significant difference in the incidence of grade 3 or higher AEs between the combined and SoC-only groups (p = 0.69). CONCLUSIONS These findings suggest that adding PD-1/PD-L1 inhibitors to standard therapy does not enhance 1-year or 2-year survival for patients with LA-HNSCC, and confers a similar severe safety profile.
Collapse
Affiliation(s)
- Long Chen
- ENT & HN Surgery Department, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, China; ENT & HN Surgery Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, China
| | - Jin-Nian He
- ENT & HN Surgery Department, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, China
| | - Shi-Jie Zhao
- ENT & HN Surgery Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, China
| | - Li-Ping Peng
- ENT & HN Surgery Department, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, China
| | - Dun-Chang Mo
- Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, China.
| | - Shi-Hua Yin
- ENT & HN Surgery Department, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, China.
| |
Collapse
|
24
|
Oppelt PJ, Ley JC, Puram SV, Jackson RS, Paniello RC, Rich JT, Pipkorn P, Liu J, Thorstad WL, Adkins DR. Palbociclib, a Selective CDK4/6 Inhibitor, Administered Before and After Chemoradiotherapy for Human Papillomavirus-Negative, Locally Advanced Head and Neck Squamous Cell Carcinoma: A Single-Arm, Phase II Trial. JCO Precis Oncol 2025; 9:e2500069. [PMID: 40403207 DOI: 10.1200/po-25-00069] [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: 01/22/2025] [Revised: 03/03/2025] [Accepted: 04/08/2025] [Indexed: 05/24/2025] Open
Abstract
PURPOSE The primary aim of this single-arm, phase II trial was to determine the objective response rate (ORR) with palbociclib, a selective CDK4/6 inhibitor, administered before chemoradiotherapy (CRT) in patients with human papillomavirus (HPV)-negative head and neck squamous cell carcinoma (HNSCC). PATIENTS AND METHODS Eligibility required untreated stage III-IVB HPV-negative HNSCC. Palbociclib 125 mg once daily was administered on days 1-21 of each 28-day cycle for two cycles before and six cycles after CRT. Pretreatment tumor samples underwent genome sequencing. The primary end point was tumor response to palbociclib given before CRT, assessed by RECIST1.1. A sample of 24 evaluable patients (those with response assessment) yielded an 80% power at a one-sided significance level of 0.05 if the ORR was ≥38% (null ORR 17%). A key secondary end point was relapse. RESULTS We enrolled 26 patients with HPV-negative, locally advanced HNSCC. Alterations in CDKN2A were identified in 15 patients (58%), and alterations in CCND1 were identified in eight (31%). Twenty-four patients were evaluable for the primary end point, and 25 were evaluable for relapse. The ORR with palbociclib given before CRT was 41.7% (95% CI, 22.1 to 63.4). Response to palbociclib occurred in 10 of 15 patients (66.7%) with versus 0 of 9 (0%) without tumor CDKN2A alterations (P = .002) and in one of six patients (16.7%) with versus nine of 18 (50%) without CCND1 alterations (P = .34). The median follow-up was 33.9 months (IQR, 31.3-49.2). Relapse occurred in two of 15 patients (13.3%) with versus seven of 10 (70%) without tumor CDKN2A alterations (P = .009) and in one of seven patients (14.3%) with versus 8 of 18 (44.4%) without CCND1 alterations (P = .36). CONCLUSION Palbociclib is an active drug in previously untreated HPV-negative, locally advanced HNSCC. Alterations in CDKN2A were associated with a higher ORR with palbociclib and a lower relapse risk after CRT.
Collapse
Affiliation(s)
- Peter J Oppelt
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO
- Alvin J. Siteman Cancer Center, St Louis, MO
- The Robert Ebert and Greg Stubblefield Head and Neck Tumor Center at Siteman, St Louis, MO
| | - Jessica C Ley
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO
| | - Sidharth V Puram
- Alvin J. Siteman Cancer Center, St Louis, MO
- The Robert Ebert and Greg Stubblefield Head and Neck Tumor Center at Siteman, St Louis, MO
- Department of Otolaryngology, Washington University School of Medicine, St Louis, MO
- Department of Genetics, Washington University School of Medicine, St Louis, MO
| | - Ryan S Jackson
- Alvin J. Siteman Cancer Center, St Louis, MO
- The Robert Ebert and Greg Stubblefield Head and Neck Tumor Center at Siteman, St Louis, MO
- Department of Otolaryngology, Washington University School of Medicine, St Louis, MO
| | - Randall C Paniello
- Alvin J. Siteman Cancer Center, St Louis, MO
- The Robert Ebert and Greg Stubblefield Head and Neck Tumor Center at Siteman, St Louis, MO
- Department of Otolaryngology, Washington University School of Medicine, St Louis, MO
| | - Jason T Rich
- Alvin J. Siteman Cancer Center, St Louis, MO
- The Robert Ebert and Greg Stubblefield Head and Neck Tumor Center at Siteman, St Louis, MO
- Department of Otolaryngology, Washington University School of Medicine, St Louis, MO
| | - Patrik Pipkorn
- Alvin J. Siteman Cancer Center, St Louis, MO
- The Robert Ebert and Greg Stubblefield Head and Neck Tumor Center at Siteman, St Louis, MO
- Department of Otolaryngology, Washington University School of Medicine, St Louis, MO
| | - Jingxia Liu
- Division of Public Health Sciences, Washington University School of Medicine, St Louis, MO
| | - Wade L Thorstad
- Alvin J. Siteman Cancer Center, St Louis, MO
- The Robert Ebert and Greg Stubblefield Head and Neck Tumor Center at Siteman, St Louis, MO
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO
| | - Douglas R Adkins
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO
- Alvin J. Siteman Cancer Center, St Louis, MO
- The Robert Ebert and Greg Stubblefield Head and Neck Tumor Center at Siteman, St Louis, MO
| |
Collapse
|
25
|
Laban S. [Medical tumor therapy for head and neck cancer: between standardization and personalization]. HNO 2025:10.1007/s00106-025-01591-7. [PMID: 40261344 DOI: 10.1007/s00106-025-01591-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2025] [Indexed: 04/24/2025]
Abstract
In the field of systemic cancer therapy for head and neck neoplasms, new approvals have led to several changes in treatment standards, primarily in the palliative situation. At the same time, molecular biomarkers have been established that can contribute to therapeutic decision-making in addition to clinical factors. These developments are summarized, and potential strategies to resolve the tension between standardization and personalization are described. Results from randomized phase III studies with therapeutic antibodies targeting the PD1/PD-L1 axis in combination with definitive chemoradiation are described and discussed. Finally, inspired by the press release about the positive results of the neoadjuvant KEYNOTE-689 trial in locoregionally advanced resectable disease, potential measures and suggestions for integrating neoadjuvant immunotherapy into clinical routine are discussed.
Collapse
Affiliation(s)
- Simon Laban
- Klinik für Hals-Nasen-Ohrenheilkunde und Kopf-Halschirurgie, Universitätsklinik Ulm, Ulm, Deutschland.
- Kopf-Hals-Tumorzentrum des Comprehensive Cancer Center Ulm, Frauensteige 12, 89070, Ulm, Deutschland.
| |
Collapse
|
26
|
Abu Taha S, Abu Hejleh T, ElHaddad M, Al-Ibraheem A, Abbasi A, Sumaida A, Bushehri A, Mostafa A, Youssef B, Alotain I, Abu-Gheida I, Aldehaim M, Alghamdi M, Shelan M, Al Dohan M, Al-Hussaini M, Pervez N, Temraz S, Alrashidi S, El-Sheshtawy W, Al-Mandhari Z, Ghatasheh H, Hosni A, Mohamad I. Chemotherapy-free innovations in locally advanced head and neck cancer: a comprehensive review. Front Oncol 2025; 15:1552337. [PMID: 40330829 PMCID: PMC12052741 DOI: 10.3389/fonc.2025.1552337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
The treatment of locally advanced head and neck squamous cell carcinoma (LA-HNSCC) has traditionally relied on a multimodal approach, combining surgery, radiation therapy (RT), and chemotherapy. While chemotherapy plays a critical role in improving cure rates and functional outcomes, its substantial toxicity remains a major concern, particularly in older patients. These challenges are especially relevant for those who are unfit for chemotherapy or decline conventional concurrent chemoradiotherapy (CCRT), highlighting the need for alternative therapeutic options. Many patients are at high risk for severe side effects, often preventing them from completing the full chemotherapy regimen. This review explores alternative strategies to definitive CCRT of carcinomas of the larynx, hypopharynx and oropharynx, aiming to optimize treatment outcomes while minimizing toxicity. We discuss altered fractionation strategies as a promising alternative to conventional RT, offering a balance between treatment efficacy and quality of life. Additionally, we examine emerging approaches, including the combining of targeted therapies, immunotherapy, hyperthermia, photodynamic therapy and nanoparticle-based treatments with RT, which provide alternative or complementary options to traditional therapies in the management of LA-HNSCC.
Collapse
Affiliation(s)
- Shatha Abu Taha
- Department of Radiation Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Taher Abu Hejleh
- Department of Medical Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Mostafa ElHaddad
- Clinical Oncology Department, Kasr Al-Ainy Center of Clinical Oncology and Nuclear Medicine, Kasr Al-Ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Akram Al-Ibraheem
- Department of Nuclear Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Ahmed Abbasi
- Department of Radiation Oncology, Aga Khan University Hospital, Karachi, Pakistan
| | | | - Ahmad Bushehri
- Department of Radiation Oncology, Kuwait Cancer Control Center, Kuwait, Kuwait
| | - Ahmad Mostafa
- Clinical oncology department, Minia Oncology, Center, Minia, Egypt
| | - Bassem Youssef
- Department of Radiation Oncology, American University of Beirut Medical Centre, Beirut, Lebanon
| | - Ibrahim Alotain
- Department of Radiation Oncology, King Fahad Specialist, Dammam, Saudi Arabia
| | - Ibrahim Abu-Gheida
- Department of Radiation Oncology, Burjeel Medical City, Abu Dhabi, United Arab Emirates
| | - Mohammed Aldehaim
- Department of Radiation Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Majed Alghamdi
- Radiation Oncology, Princess Noorah Oncology Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs-Western Region, Jeddah, Saudi Arabia
- Collage of Medicine, King Saud Bin Abdulaziz University for Health Science, Jeddah, Saudi Arabia
| | - Mohamed Shelan
- Department of Radiation Oncology, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Mohammed Al Dohan
- Department of Radiation Oncology, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Maysa Al-Hussaini
- Department of Cell Therapy and Applied Genomics, King Hussein Cancer Center, Amman, Jordan
| | - Nadeem Pervez
- Department of Radiation Oncology, United Arab Emirates (UAE) University, Al Ain, United Arab Emirates
| | - Shoukri Temraz
- Clinical Oncology Department, Mansoura University hospital, Mansour, Egypt
| | - Saad Alrashidi
- Department of Radiation Oncology, Comprehensive Cancer Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | | | - Zahid Al-Mandhari
- Department of Radiation Oncology, Sultan Qaboos Comprehensive Cancer Care and Research Centre, Muscat, Oman
| | - Hamza Ghatasheh
- Department of Radiation Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Ali Hosni
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Issa Mohamad
- Department of Radiation Oncology, King Hussein Cancer Center, Amman, Jordan
| |
Collapse
|
27
|
Cavalieri S, Brakenhoff RH, Leemans CR, Hoebers FJP, Poli T, Scheckenbach K, Iacovelli NA, Franceschini M, Orlandi E, Licitra L, De Cecco L. Prognostic gene expression signatures for HPV-negative head and neck squamous cell carcinoma. Radiother Oncol 2025; 208:110900. [PMID: 40252811 DOI: 10.1016/j.radonc.2025.110900] [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: 10/19/2024] [Revised: 03/24/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is a leading cause of cancer-related deaths worldwide, with HPV-negative cases being particularly aggressive. These cases often show poor prognosis and low responsiveness to radiotherapy. Improved prognostic tools and treatment strategies are needed to enhance outcomes. AIM To evaluate the prognostic value of various gene expression signatures in predicting survival outcomes in HPV-negative HNSCC patients receiving radiotherapy and to compare their accuracy against the current TNM staging system. METHODS This observational cohort study used data from the European BD2Decide project, systematically analyzing gene expression in loco-regionally advanced, non-metastatic HPV-negative HNSCC patients (stage III-IVa/b) treated with curative radiotherapy (post-operative or definitive) between 2008 and 2017. The primary outcome was overall survival (OS), with secondary outcomes including disease-free survival (DFS), distant metastasis-free survival (DMFS), and loco-regional recurrence-free survival (LRRFS). The prognostic performance of selected gene expression signatures was evaluated using receiver operating characteristic (ROC) curves and hazard ratios (HR) from Cox models. RESULTS The study included 783 patients, with a median age of 63 years, mostly male (68 %), with significant tobacco (84 %) and alcohol (69 %) exposure. The 172-gene signature (172GS) showed the highest prognostic accuracy, outperforming the TNM system in predicting OS, DFS, DMFS, and LRRFS. Multivariable analysis confirmed its independent prognostic value. CONCLUSIONS The 172GS gene signature offers superior prognostic information compared to TNM staging, supporting its potential use for better risk stratification and personalized treatment planning in HPV-negative HNSCC. Future trials should consider tumor biology and gene signatures for better patient selection. TRIAL REGISTRATION NCT02832102.
Collapse
Affiliation(s)
- Stefano Cavalieri
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy; Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy.
| | - Ruud H Brakenhoff
- Amsterdam UMC location Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, De Boelelaan 1117, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands
| | - C René Leemans
- Amsterdam UMC location Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, De Boelelaan 1117, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands
| | - Frank J P Hoebers
- Department of Radiation Oncology (MAASTRO), Maastricht University GROW School for Oncology and Reproduction, MAASTRO Clinic, Maastricht, the Netherlands
| | - Tito Poli
- Department of Medicine and Surgery, University of Parma & Head & Neck Department, University Hospital of Parma, Parma, Italy
| | - Kathrin Scheckenbach
- Department of Otolaryngology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | | | - Marzia Franceschini
- Radiotherapy Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Ester Orlandi
- Radiotherapy Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy; Radiation Oncology Clinical Department, National Center for Oncological Hadron Therapy (CNAO), Pavia, Italy; Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Lisa Licitra
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy; Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Loris De Cecco
- Integrated Biology of Rare Tumors, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
28
|
Timnik VR, Zoeschg A, Diederich S, Nefzger SM, Huang Z, Schmid NA, Giller M, Steiger K, Combs SE, Kroemer G, Schmid TE, Fischer JC. Experimental Investigation of Hematological Toxicity After Radiation Therapy Combined With Immune Checkpoint Inhibitors. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00372-4. [PMID: 40250771 DOI: 10.1016/j.ijrobp.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/28/2025] [Accepted: 04/05/2025] [Indexed: 04/20/2025]
Abstract
PURPOSE Combining immune checkpoint inhibitors (ICIs) with radiation therapy (RT) has led to significant advancements in cancer treatment. However, evidence from clinical and experimental studies suggests that this combination may increase hematopoietic and lymphatic toxicity. This study aims to investigate the effects of the concurrent application of ICIs (anti-PD-1 and anti-CTLA-4) on radiation-induced hematopoietic and lymphatic injuries under standardized and controlled experimental conditions. METHODS AND MATERIALS We used various experimental models in C57BL/6 and BALB/c mice to evaluate the impact of ICIs combined with RT on the hematopoietic system. These models involved different RT doses, regimens, and target sites in both healthy and tumor-bearing mice. RESULTS Our findings showed that the concurrent use of ICIs did not meaningfully affect post-RT pancytopenia kinetics or the regeneration of specific blood cell lineages over time. Consistently, combining RT with ICIs did not significantly enhance DNA damage in immune cells within the bloodstream. This outcome was comparable across different RT doses, regimens, and target sites and was reproducible in both tumor-bearing and nontumor-bearing mice. Additionally, there were no significant increases in late side effects, including reductions in bone marrow cell counts or megakaryocyte numbers, after combined radioimmunotherapy. CONCLUSIONS These findings suggest that combining ICIs with RT does not exacerbate hematological toxicity. This information is valuable for interpreting adverse events in clinical trials involving radioimmunotherapy and for predicting potential hematological side effects in cancer patients receiving these treatments.
Collapse
Affiliation(s)
- Vincent R Timnik
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Andreas Zoeschg
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Sarah Diederich
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Sophie M Nefzger
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Ziyi Huang
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Nicole A Schmid
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Maximilian Giller
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Katja Steiger
- Comparative Experimental Pathology (CEP), Institute of Pathology, School of Medicine, Technical University of Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany; Helmholtz Zentrum München, Institute of Radiation Medicine, Neuherberg, Germany
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, France-HP, Paris, France
| | - Thomas E Schmid
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany; Helmholtz Zentrum München, Institute of Radiation Medicine, Neuherberg, Germany
| | - Julius C Fischer
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France.
| |
Collapse
|
29
|
Liu X, Harbison RA, Varvares MA, Puram SV, Peng G. Immunotherapeutic strategies in head and neck cancer: challenges and opportunities. J Clin Invest 2025; 135:e188128. [PMID: 40231472 PMCID: PMC11996880 DOI: 10.1172/jci188128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025] Open
Abstract
HNSCC remains a substantial health issue, with treatment options including surgery, radiation, and platinum-based chemotherapy. Unfortunately, despite progress in research, only modest gains have been made in disease control, with existing treatments resulting in significant functional and quality-of-life issues. The introduction of immunotherapy in the treatment of HNSCC has resulted in some improvements in outlook for patients and is now standard of care for populations with both recurrent and metastatic disease. However, despite the early successes, responses to immune checkpoint inhibition (ICI) remain modest to low, approaching 14%-22% objective response rates. Challenges to the effectiveness of ICI and other immunotherapies are complex, including the diverse and dynamic molecular plasticity and heterogeneity of HNSCCs; lack of immunogenic antigens; accumulated suppressive immune populations such as myeloid cells and dysfunctional T cells; nutrient depletion; and metabolic dysregulation in the HNSCC tumor microenvironment. In this Review, we explore the mechanisms responsible for immunotherapy resistance, dissect these challenges, and discuss potential opportunities for overcoming hurdles to the development of successful immunotherapy for HNSCC.
Collapse
Affiliation(s)
- Xia Liu
- Department of Otolaryngology–Head and Neck Surgery
- Rob Ebert and Greg Stubblefield Head and Neck Tumor Center at Siteman Cancer Center and
| | - R. Alex Harbison
- Department of Otolaryngology–Head and Neck Surgery
- Rob Ebert and Greg Stubblefield Head and Neck Tumor Center at Siteman Cancer Center and
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Mark A. Varvares
- Department of Otolaryngology–Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - Sidharth V. Puram
- Department of Otolaryngology–Head and Neck Surgery
- Rob Ebert and Greg Stubblefield Head and Neck Tumor Center at Siteman Cancer Center and
- Department of Genetics, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Guangyong Peng
- Department of Otolaryngology–Head and Neck Surgery
- Rob Ebert and Greg Stubblefield Head and Neck Tumor Center at Siteman Cancer Center and
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
30
|
Shen Y, Connolly E, Aiello M, Zhou C, Chappa P, Song H, Tippitak P, Clark T, Cardenas M, Prokhnevska N, Mariniello A, De Bruyker I, Pagadala MS, Dhere VR, Rafiq S, Kesarwala AH, Orthwein A, Thomas SN, Zhang SL, Khan MK, Dixon JB, Lesinski GB, Lowe MC, Kissick H, Yu DS, Paulos CM, Schmitt NC, Buchwald ZS. Combination radiation and αPD-L1 enhance tumor control by stimulating CD8+ PD-1+ TCF-1+ T cells in the tumor-draining lymph node. Nat Commun 2025; 16:3522. [PMID: 40229241 PMCID: PMC11997041 DOI: 10.1038/s41467-025-58510-1] [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: 02/12/2024] [Accepted: 03/19/2025] [Indexed: 04/16/2025] Open
Abstract
Combination radiotherapy (RT) and αPD-L1 therapy has potential to enhance local and distant (abscopal) tumor control, however, clinical results in humans have been variable. Using murine melanoma models, we found RT + αPD-L1 increases intra-tumor progenitor CD8+ PD-1+ TCF-1+ T cells. This increase depends on trafficking of the PD-1+ TCF-1+ cells from the tumor-draining lymph node (TdLN) to the tumor. RT alone promotes the expansion and differentiation of the TdLN derived PD-1+ TCF-1+ cells into TIM-3+ GZMB+ TCF-1- effector-like cells in the tumor with further enhancement after the addition of αPD-L1. In the TdLN, combination therapy enriches for a novel PD-1+ TCF-1+ TOX- LY6A+ subset with expression of a type I interferon and migratory signature. This subset is able to traffic to the tumor and differentiate into TIM-3+ TCF-1- cells. Finally, we found that ablation of the PD-1+ TCF-1+ T cell population attenuates the enhanced tumor control observed with combination RT + αPD-L1. These results suggest that abscopal response failures may be secondary to impaired stimulation of TdLN CD8+ PD-1 + TCF-1+ T cells or an inability of PD-1+ TCF-1+ cells in the TdLN to traffic to the tumor.
Collapse
Affiliation(s)
- Yang Shen
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Erin Connolly
- Bioinformatics Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA
| | - Meili Aiello
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Chengjing Zhou
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Prasanthi Chappa
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Haorui Song
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Patan Tippitak
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Tarralyn Clark
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Maria Cardenas
- Department of Urology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Nataliya Prokhnevska
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai (ICMMS), New York City, NY, USA
| | - Annapaola Mariniello
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Isabelle De Bruyker
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Meghana S Pagadala
- Medical Scientist Training Program, University of California San Diego, La Jolla, CA, USA
| | - Vishal R Dhere
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Sarwish Rafiq
- Department of Hematology and Medical Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Aparna H Kesarwala
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Alexandre Orthwein
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Susan N Thomas
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Shirley L Zhang
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mohammad K Khan
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - J Brandon Dixon
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Gregory B Lesinski
- Department of Hematology and Medical Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Michael C Lowe
- Department of Surgery and Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Haydn Kissick
- Department of Urology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - David S Yu
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Chrystal M Paulos
- Department of Surgery and Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Nicole C Schmitt
- Department of Otolaryngology - Head and Neck Surgery and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Zachary S Buchwald
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| |
Collapse
|
31
|
Saenz FR, Velasquez B, Waldrop T, Aguilar E, Cox KR, Delahoussaye A, Laberiano-Fernandez C, Clemente LC, Connell L, Mims N, Neill D, Parra ER, Clise-Dwyer K, Schüler E, Spiotto MT. FLASH radiotherapy spares lymphocytes in tumor-draining lymph nodes and increases infiltration of immune cells in tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.07.647544. [PMID: 40291670 PMCID: PMC12026895 DOI: 10.1101/2025.04.07.647544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Radiotherapy (RT) delivered at conventional dose rates (CONV) can both stimulate antitumor immune responses and inhibit these immune responses by depleting circulating lymphocytes. Given the observed normal tissue sparing associated with ultra-high dose rate (FLASH) RT, we hypothesized that FLASH RT may protect lymphocytes while increasing the immunogenicity of cancer cells. We irradiated cancer cell lines in vitro with FLASH RT or CONV RT and assessed immunogenic mRNA and protein expression. Both HPV-positive cell lines MEER and TC-1 showed upregulation of Calr, Hmgb1 , and cGAS-STING family members after FLASH RT but not after CONV RT in vitro . To assess changes in lymphocyte populations, we irradiated murine mEER tumors in syngeneic C57BL/6 mice with 27 Gy in 3 fractions of FLASH RT or CONV RT. In mice bearing FLASH irradiated tumors, tumor-draining lymph nodes contained greater numbers of CD8 + T cells (FLASH 1.7×10 4 vs 0.8×10 4 CONV; P <0.001) and CD4 + T cells (FLASH 2.3×10 4 vs CONV 1.2×10 4 ; P <0.001) after irradiation. FLASH RT was associated with increased numbers of activated CD44 + CD62L lo CD8 + and CD4 + lymphocytes. In irradiated tumors, FLASH RT was associated with increased CD8 + tumor-infiltrating lymphocytes, increased PD1 expression on these lymphocytes and increased PDL1 expression on macrophages. Compared with CONV RT, FLASH RT spared activated T cells in tumor-draining lymph nodes and in tumors but increased checkpoint inhibitor expression in tumors. These results suggest that FLASH RT may enhance antitumor immune responses by maintaining the immunogenic effects of RT while preserving lymphocyte numbers, which may be augmented with immune checkpoint blockade. Significance Radiation-induced lymphopenia is associated with poorer survival outcomes. New treatment approaches, like FLASH radiation therapy (FLASH RT), which reduce lymphopenia and enhance the antitumor response, could potentially lead to better outcomes for cancer patients.
Collapse
|
32
|
Li Y, Yadollahi P, Essien FN, Putluri V, Ambati CSR, Kami Reddy KR, Kamal AHM, Putluri N, Abdurrahman LM, Ruiz Echartea ME, Ernste KJ, Trivedi AJ, Vazquez-Perez J, Hudson WH, Decker WK, Patel R, Osman AA, Kheradmand F, Lai SY, Myers JN, Skinner HD, Coarfa C, Lee K, Jain A, Malovannaya A, Frederick MJ, Sandulache VC. Tobacco smoke exposure is a driver of altered oxidative stress response and immunity in head and neck cancer. J Transl Med 2025; 23:403. [PMID: 40188338 PMCID: PMC11971752 DOI: 10.1186/s12967-025-06258-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/17/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Exposomes are critical drivers of carcinogenesis. However, how they modulate tumor behavior remains unclear. Extensive clinical data show cigarette smoke to be a key exposome that promotes aggressive tumors, higher rates of metastasis, reduced response to chemoradiotherapy, and suppressed anti-tumor immunity. We sought to determine whether smoke itself can modulate aggressive tumor behavior in head and neck squamous cell carcinoma (HNSCC) through reprogramming of the cellular reductive state. METHODS Using established human and murine HNSCC cell lines and syngeneic mouse models, we utilized conventional western blotting, steady state and flux metabolomics, RNA sequencing, quantitative proteomics and flow cytometry to analyze the impact of smoke exposure on HNSCC tumor biology and anti-tumor immunity. RESULTS Cigarette smoke persistently activated Nrf2 target genes essential for maintenance of the cellular reductive state and survival under conditions of increased oxidative stress in HNSCC regardless of human papillomavirus (HPV) association. In contrast to e-cigarette vapor, conventional cigarette smoke mobilizes cellular metabolism toward oxidative stress adaptation, resulting in development of cross-resistance to cisplatin. In parallel, smoke exposure modulates expression of PDL1 and the secretory phenotype of HNSCC cells resulting in an altered tumor immune microenvironment (TIME) in syngeneic mouse models and downregulated expression of antigen presentation and costimulatory genes in myeloid cells. CONCLUSION The cigarette smoke exposome is a potent activator of the Nrf2 pathway and appears to be the primary trigger for a tripartite phenotype of aggressive HNSCC consisting of: (1) reduced chemotherapy sensitivity, (2) enhanced metastatic potential and (3) suppressed anti-tumor immunity.
Collapse
Affiliation(s)
- Yang Li
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, One Baylor Plaza, MS: NA102, Houston, TX, 77030, USA
| | - Pedram Yadollahi
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, One Baylor Plaza, MS: NA102, Houston, TX, 77030, USA
| | - Fonma N Essien
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, One Baylor Plaza, MS: NA102, Houston, TX, 77030, USA
| | - Vasanta Putluri
- Advanced Technology Cores, Dan Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Chandra Shekar R Ambati
- Advanced Technology Cores, Dan Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Karthik Reddy Kami Reddy
- Advanced Technology Cores, Dan Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Abu Hena Mostafa Kamal
- Advanced Technology Cores, Dan Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Nagireddy Putluri
- Advanced Technology Cores, Dan Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Lama M Abdurrahman
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, One Baylor Plaza, MS: NA102, Houston, TX, 77030, USA
| | - Maria E Ruiz Echartea
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Keenan J Ernste
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Akshar J Trivedi
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | | | - William H Hudson
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Center for Cell Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - William K Decker
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Center for Cell Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Rutulkumar Patel
- Department of Radiation Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Abdullah A Osman
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farrah Kheradmand
- Department of Medicine-Pulmonary, Baylor College of Medicine, Houston, TX, USA
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Stephen Y Lai
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Jeffrey N Myers
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heath D Skinner
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cristian Coarfa
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Kwangwon Lee
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA
| | - Antrix Jain
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA
| | - Anna Malovannaya
- Verna and Marrs Mclean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA
| | - Mitchell J Frederick
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, One Baylor Plaza, MS: NA102, Houston, TX, 77030, USA
| | - Vlad C Sandulache
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, One Baylor Plaza, MS: NA102, Houston, TX, 77030, USA.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA.
| |
Collapse
|
33
|
Price JM, Mell LK. Managing Patients with Head and Neck Cancer and Advanced Age or Comorbidities. Semin Radiat Oncol 2025; 35:197-206. [PMID: 40090746 DOI: 10.1016/j.semradonc.2025.02.001] [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: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 03/18/2025]
Abstract
The dominant treatment paradigm for locoregionally advanced head and neck squamous cell carcinoma (HNSCC) involves postoperative or definitive radiotherapy with concurrent cisplatin chemotherapy. Despite years of research investigating de-intensified treatment, cisplatin-based chemoradiotherapy remains the standard, yet it is associated with significant acute and chronic toxicity. However, due to shared risk factors, such as advanced age, and tobacco and alcohol use, patients with HNSCC frequently have comorbid illnesses that impact treatment tolerability, adding complexity to treatment-related decision-making. In addition, many patients have medical contraindications to cisplatin, requiring alternative treatment strategies. It is thus important to consider how well patients are likely to tolerate treatment, and how to adapt treatment in response to a patient's condition, when weighing treatment options. In this review, we aim to offer readers guidance in managing the elderly or comorbid patient with HNSCC, with particular attention to (i) approaching comorbidity and fragility assessment to make determinations on intensity of treatment, (ii) considering primary treatment modality (eg, surgery vs radiotherapy, chemo-radiotherapy vs radiotherapy alone) and (iii) choice of concurrent systemic therapy agent.
Collapse
Affiliation(s)
- James M Price
- The Christie NHS Foundation Trust, Manchester, UK; The University of Manchester, Manchester, UK
| | - Loren K Mell
- Department of Radiation Medicine & Applied Sciences, University of California San Diego, La Jolla, CA; Gleiberman Head and Neck Cancer Center, La Jolla, CA.
| |
Collapse
|
34
|
Trada Y, Lee MT, Jameson MG, Chlap P, Keall P, Moses D, Lin P, Fowler A. Mid-treatment changes in intra-tumoural metabolic heterogeneity correlate to outcomes in oropharyngeal squamous cell carcinoma patients. EJNMMI Res 2025; 15:31. [PMID: 40167887 PMCID: PMC11961835 DOI: 10.1186/s13550-025-01226-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/12/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND This study evaluated mid-treatment changes in intra-tumoural metabolic heterogeneity and quantitative FDG-PET/CT imaging parameters and correlated the changes with treatment outcomes in oropharyngeal squamous cell cancer (OPSCC) patients. 114 patients from two independent cohorts underwent baseline and mid-treatment (week 3) FDG-PET. Standardized uptake value maximum (SUVmax), standardized uptake value mean (SUVmean), metabolic tumour volume (MTV), and total lesional glycolysis (TLG) were measured. Intra-tumoural metabolic heterogeneity was quantified as the area under a cumulative SUV-volume histogram curve (AUC-CSH). Baseline and relative change (%∆) in imaging features were correlated to locoregional recurrence free survival (LRRFS) using multivariate Cox regression analysis. Patients were stratified into three risk groups utilising ∆AUC-CSH and known prognostic features, then compared using Kaplan-Meier analysis. RESULTS Median follow up was 39 months. 18% of patients developed locoregional recurrence at 2 years. A decrease in heterogeneity (∆AUC-CSH: 24%) was observed mid-treatment. There was no statistically significant difference in tumour heterogeneity (AUC-CSH) at baseline (p = 0.134) and change at week 3 (p = 0.306) between p16 positive and p16 negative patients. Baseline imaging features did not correlate to LRRFS. However, ∆MTV (aHR 1.04; 95% CI 1.03-1.06; p < 0.001) and ∆AUC-CSH (aHR 0.96; 95% CI 0.94-0.98; p = 0.004) were correlated to LRRFS. Stratification using ∆AUC-CSH and p16 status into three groups showed significant differences in LRR (2 year LRRFS 94%, 79%, 17%; log rank p < 0.001). Stratification using ∆AUC-CSH and ∆MTV into three groups showed significant differences in LRR (2 year LRRFS 93%, 70%, 17%; log rank p < 0.001). CONCLUSION Mid-treatment changes in intra-tumoural FDG-PET/CT heterogeneity correlated with treatment outcomes in OPSCC and may help with response prediction. These findings suggest potential utility in designing future risk adaptive clinical trials.
Collapse
Affiliation(s)
- Yuvnik Trada
- Department of Radiation Oncology, Calvary Mater Newcastle, Edith St, Waratah, 2298, NSW, Australia.
- Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.
| | - Mark T Lee
- Department of Radiation Oncology, Cancer Therapy Centre, Liverpool Hospital, Liverpool, NSW, Australia
- South Western Clinical School, School of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Michael G Jameson
- GenesisCare, Sydney, NSW, Australia
- St Vincent's clinical school, Faculty of Medicine, University NSW, Sydney, Australia
| | - Phillip Chlap
- Department of Radiation Oncology, Cancer Therapy Centre, Liverpool Hospital, Liverpool, NSW, Australia
- South Western Clinical School, School of Medicine, University of New South Wales, Sydney, NSW, Australia
- Ingham Institute of Applied Medical Research, Liverpool, NSW, Australia
| | - Paul Keall
- Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
- Image X Institute, University of Sydney, Sydney, NSW, Australia
| | - Daniel Moses
- Graduate school of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, Australia
- Department of Medical Imaging, Prince of Wales Hospital, Randwick, NSW, Australia
| | - Peter Lin
- South Western Clinical School, School of Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Nuclear Medicine and PET, Liverpool Hospital, Liverpool, NSW, Australia
- School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Allan Fowler
- Department of Radiation Oncology, Cancer Therapy Centre, Liverpool Hospital, Liverpool, NSW, Australia
- South Western Clinical School, School of Medicine, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
35
|
Safavi AH, Lee NY, Tsai CJ. Principles and Paradigms of De-Escalated Elective Nodal Irradiation: Boldly Going Towards an Inflection Point in Head and Neck Radiotherapy. Semin Radiat Oncol 2025; 35:143-156. [PMID: 40090741 DOI: 10.1016/j.semradonc.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 01/16/2025] [Indexed: 03/18/2025]
Abstract
Fundamental axioms of elective nodal irradiation (ENI) for head and neck cancers merit re-examination in contemporary practice. Standard ENI doses to volumes bordering critical organs-at-risk increased during the transition from two-dimensional radiation planning to intensity-modulated radiotherapy, despite improvements in detection of occult nodal metastases with modern imaging, use of concurrent chemotherapy, and identification of human papillomavirus (HPV)-related radiosensitivity. Historical large ENI volumes covering low-risk nodal regions continue to be commonly used even as awareness grows regarding the predominant pattern-of-failure within existing gross disease. In this review, we outline principles for de-escalating head and neck ENI dose and volume and highlight the emerging paradigm of ENI omission. We also propose a three-part approach to ENI de-escalation, the rationale for early adoption of de-escalated ENI in the absence of level-one evidence, and strategies to promote early adoption in light of modest equipoise and an inflection point towards changing the status quo.
Collapse
Affiliation(s)
- Amir H Safavi
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada.; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nancy Y Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - C Jillian Tsai
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada.; Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada..
| |
Collapse
|
36
|
Beier J, Rühle A. [Definitive radiotherapy with cetuximab or durvalumab for locoregionally advanced head and neck cancer in patients with a contraindication to cisplatin (NRG-HN004)]. Strahlenther Onkol 2025; 201:475-477. [PMID: 39904780 DOI: 10.1007/s00066-025-02370-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2025] [Indexed: 02/06/2025]
Affiliation(s)
- Josephine Beier
- Klinik und Poliklinik für Strahlentherapie, Universitätsklinikum Leipzig, Stephanstraße 9a, 04103, Leipzig, Deutschland
- Partnerstandort Leipzig, Mitteldeutsches Krebszentrum (CCCG), Leipzig, Deutschland
| | - Alexander Rühle
- Klinik und Poliklinik für Strahlentherapie, Universitätsklinikum Leipzig, Stephanstraße 9a, 04103, Leipzig, Deutschland.
- Partnerstandort Leipzig, Mitteldeutsches Krebszentrum (CCCG), Leipzig, Deutschland.
| |
Collapse
|
37
|
Widjaja W, Ng I, Shannon N, Iyer NG. Neoadjuvant Immunotherapy in Resectable HNSCC: An Updated Systematic Review and Meta-analysis. Ann Surg Oncol 2025; 32:2713-2724. [PMID: 39645552 DOI: 10.1245/s10434-024-16587-w] [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: 07/24/2024] [Accepted: 11/12/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Immunotherapy is a recently recognised FDA-approved treatment for R/M HNSCC. Our goal is to explore the safety profile and the efficacy of immunotherapy in the neoadjuvant setting before surgery in mucosal head and neck cancer. METHODS Three electronic databases had been systematically searched through March 2024. Demographic and tumour characteristics were extracted. Primary outcomes obtained were disease-free survival (DFS), progression-free survival (PFS), overall survival (OS), complete pathological response (cPR), which was defined as no residual tumour, and major pathological response (MPR), which as defined as <10% residual viable tumour. Safety outcomes examined were grade 3 and above adverse event, median time to surgery, delays to surgery, and death related to neoadjuvant treatment. RESULTS A total of 459 patients from 15 studies were included in the analysis. The pooled estimate of cPR for all the studies was 14.9% (95% confidence interval [CI] 8.0-26.2). Subgroup analysis showed chemoimmunotherapy had a higher cPR 30.1% (95% CI 22.8-38.62) compared with immunotherapy alone 1.4% (95% CI 0.3-5.2). There was no treatment-related death. Chemoimmunotherapy had a higher pooled estimate of adverse events 22.9% (95% CI 11.0-41.5) compared with immunotherapy alone 8.5% (95% CI 2.6-24.3). Subgroup analysis demonstrated that chemoimmunotherapy had a higher DFS compared with immunotherapy alone: 89.8% (95% CI 81.4-94.7) versus 80.44% (95% CI 73.9-85.7), respectively. Neoadjuvant immunoradiotherapy had conflicting results. CONCLUSIONS Neoadjuvant immunotherapy was well tolerated. Neoadjuvant chemoimmunotherapy may be more effective in treating LAHNSCC over immunotherapy alone; however, TRAEs were higher.
Collapse
Affiliation(s)
- Winy Widjaja
- Department of Head and Neck Surgery, National Cancer Centre Singapore, Singapore, Singapore.
| | - Irene Ng
- Department of Head and Neck Surgery, National Cancer Centre Singapore, Singapore, Singapore
| | - Nicolas Shannon
- Department of Head and Neck Surgery, National Cancer Centre Singapore, Singapore, Singapore
| | - N Gopalakrishna Iyer
- Department of Head and Neck Surgery, National Cancer Centre Singapore, Singapore, Singapore
| |
Collapse
|
38
|
Ma D, Routman DM. De-escalation of Adjuvant Therapy in Operatively Managed HPV Associated Oropharyngeal Carcinoma: Current Status and Future Directions. Semin Radiat Oncol 2025; 35:166-172. [PMID: 40090743 DOI: 10.1016/j.semradonc.2025.02.002] [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: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 03/18/2025]
Abstract
Human papillomavirus (HPV) associated oropharyngeal carcinoma is currently the most frequently diagnosed head and neck cancer in the United States. Due to the generally high cure rates with standard therapies, de-intensification strategies are being explored to reduce acute and long-term side effects. For patients treated with definitive chemoradiation, unselected de-escalation has shown worse progression-free survival compared to standard therapy. Concurrently, surgical management is becoming more prevalent, and adjuvant de-escalation appears promising. Further research is required to identify optimal candidacy for adjuvant de-escalation and to understand the relationship between dose and volume de-escalation. Biomarkers such as ctDNA may assist in candidate selection, but validation and alignment with pathological criteria are necessary.
Collapse
Affiliation(s)
- Daniel Ma
- Department of Radiation Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, MN..
| | - David M Routman
- Department of Radiation Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, MN
| |
Collapse
|
39
|
Hill J, Schoenfeld JD. Immunotherapy With Curative Intent Radiotherapy for Patients With Cancers of the Head and Neck. Semin Radiat Oncol 2025; 35:214-223. [PMID: 40090748 DOI: 10.1016/j.semradonc.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/15/2025] [Accepted: 02/16/2025] [Indexed: 03/18/2025]
Affiliation(s)
- Jordan Hill
- Banner MD Anderson Cancer Center, Gilbert, AZ.
| | | |
Collapse
|
40
|
Hasibuzzaman MM, He R, Khan IN, Salem AK, Simons AL. Radiotherapy is enhanced by CPH:SA IL-1α microparticles in a murine HNSCC tumor model. BMC Cancer 2025; 25:588. [PMID: 40169985 PMCID: PMC11963532 DOI: 10.1186/s12885-025-13995-3] [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: 01/21/2025] [Accepted: 03/24/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Radiotherapy (RT) can trigger immunogenic cell death which may be exploited to improve the effectiveness of immunotherapy. However, recent results from clinical trials testing RT/immunotherapy combinations in head and neck squamous cell carcinoma patients (HNSCC) have been disappointing. Interleukin-1 alpha (IL-1α) is a cytokine that can activate various aspects of anti-tumor immunity including dendritic cell (DC) activation which is critical for the recruitment of tumor infiltrating lymphocytes. Here we test the cytokine IL-1α encapsulated in 20:80 1,6-bis-(p-carboxyphenoxy)-hexane:sebacic acid (CPH:SA) copolymer-based microparticles (IL-1αMPs) as an adjuvant to RT in a murine syngeneic HNSCC mouse model. Thus the main research objective of this current study was to evaluate if IL-1αMPs can enhance the antitumor immune response of radiotherapy. METHODS Activation of immune cells in response to RT ± human recombinant IL-1α was evaluated in human peripheral blood mononuclear cell (PBMC):cancer cell co-cultures. A bilateral HNSCC tumor syngeneic mouse model was used to monitor mEERL tumor growth and immune cell recruitment in response to RT (8 Gy to irradiated tumor only) with and without intraperitoneal delivery of IL-1αMPs. RESULTS: Results showed that IL-1α induced the activation of monocytes, NK cells, T cells, and DCs in PBMC:Cal-27 cell co-cultures but there was no enhanced immune cell activation (with the exception of NK cells) in vitro when combined with RT. RT and RT + IL-1αMPs significantly suppressed growth in irradiated mEERL tumors compared to control. However, only the combination therapy was able to slowdown growth of the non-irradiated tumors compared to the other treatment groups. Immune cell profiling revealed that RT caused acute lymphodepletion on treatment day 3 which was reversed by treatment day 11 in RT-exposed mice. The anti-tumor effect of RT + IL-1α was accompanied by significantly increased infiltration of DCs in the irradiated tumor and increased CD8 + and antigen (E7)-specific CD8 + T cell infiltration in both irradiated and non-irradiated tumors. The anti-tumor response of the combination therapy was completely abrogated by CD8 + T cell depletion. CONCLUSIONS This data suggests that the addition of CPH:SA IL-1αMPs to RT may boost anti-tumor immune response and target both local and systemic disease. This combination is worthy of further investigation as an immunotherapeutic strategy and could represent a promising approach to improve survival outcomes in HNSCC patients.
Collapse
Affiliation(s)
- M M Hasibuzzaman
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, USA
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa Hospital and Clinics, University of Iowa, Iowa City, IA, USA
| | - Rui He
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
| | - Ishrat Nourin Khan
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, USA
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa Hospital and Clinics, University of Iowa, Iowa City, IA, USA
| | - Aliasger K Salem
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa Hospital and Clinics, University of Iowa, Iowa City, IA, USA
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
| | - Andrean L Simons
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, USA.
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, University of Iowa, Iowa City, IA, USA.
- Holden Comprehensive Cancer Center, University of Iowa Hospital and Clinics, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
41
|
Cao LM, Yu YF, Li ZZ, Wang GR, Xiao Y, Luo HY, Liu B, Bu LL. Neoadjuvant Chemoimmunotherapy for Resectable Head and Neck Squamous Cell Carcinoma: Systematic Review and Meta-analysis. Ann Surg Oncol 2025:10.1245/s10434-025-17195-y. [PMID: 40102288 DOI: 10.1245/s10434-025-17195-y] [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: 12/05/2024] [Accepted: 03/03/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Neoadjuvant chemoimmunotherapy provides a good pathological response in patients with resectable head and neck squamous cell carcinoma (HNSCC). Currently, there is no comprehensive systematic review that rigorously evaluates and summarizes the existing studies. In this study, we aimed to synthesize the results on the efficacy of neoadjuvant chemoimmunotherapy in resectable HNSCC to obtain higher-level evidence. METHODS The PubMed, Web of Science, Scopus, and Academic Search Complete (EBSCO) databases, along with ClinicalTrials.gov, Google Scholar, and conference abstracts, were comprehensively searched. The publication dates of the literature were limited to January 2015-July 2024. Meta-analysis was performed using a random-effects model. The percentage of major pathological response (MPR), pathological complete response (pCR), and overall disease-free survival (DFS) were synthesized. The odds ratios of a combined positive score (CPS) ≥ 20 for MPR and the diagnostic performance of using radiological objective response to determine MPR were further explored. RESULTS A total of 13 studies with 458 patients who received neoadjuvant chemoimmunotherapy and 443 patients who underwent curative surgery were included. The pooled MPR, pCR, and overall DFS rates were 61%, 37%, and 91%, respectively. The odds ratios of a CPS ≥ 20 for achieving MPR was 2.09 compared with those with a CPS < 20. The sensitivity of using radiological objective response to determine MPR was 0.91 and the specificity was 0.46, with an area under the curve of 0.76. CONCLUSION Neoadjuvant chemoimmunotherapy showed promising results for resectable HNSCC. A CPS ≥ 20 can be used to screen for treatment-sensitive patients, and radiological examinations can be used to detect pathological response. Definitive conclusions require data from longer follow-up periods and controlled studies.
Collapse
Affiliation(s)
- Lei-Ming Cao
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yi-Fu Yu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yao Xiao
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Han-Yue Luo
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Bing Liu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
42
|
Sakai SA, Saeki K, Chi S, Hamaya Y, Du J, Nakamura M, Hojo H, Kojima T, Nakamura Y, Bando H, Kojima M, Suzuki A, Suzuki Y, Akimoto T, Tsuchihara K, Haeno H, Yamashita R, Kageyama SI. Mathematical Modeling Predicts Optimal Immune Checkpoint Inhibitor and Radiotherapy Combinations and Timing of Administration. Cancer Immunol Res 2025; 13:353-364. [PMID: 39666379 PMCID: PMC11876959 DOI: 10.1158/2326-6066.cir-24-0610] [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: 07/02/2024] [Revised: 09/04/2024] [Accepted: 12/10/2024] [Indexed: 12/13/2024]
Abstract
Radiotherapy (RT) combined with immune checkpoint inhibitor (ICI) therapy has attracted substantial attention due to its potential to improve outcomes for patients with several types of cancer. However, the optimal administration timepoints and drug combinations remain unclear because the mechanisms underlying RT-induced changes in immune checkpoint molecule expression and interaction with their ligand(s) remain unclear. In this study, we demonstrated the dynamics of lymphocyte-mediated molecular interactions in tissue samples from patients with esophageal cancer throughout RT schedules. Single-cell RNA sequencing and spatial transcriptomic analyses were performed to investigate the dynamics of these interactions. The biological signal in lymphocytes transitioned from innate to adaptive immune reaction, with increases in ligand-receptor interactions, such as PD-1-PD-L1, CTLA4-CD80/86, and TIGIT-PVR interactions. A mathematical model was constructed to predict the efficacy of five types of ICIs when administered at four different timepoints. The model suggested that concurrent anti-PD-1/PD-L1 therapy or concurrent/adjuvant anti-CTLA4/TIGIT therapy would exert a maximal effect with RT. This study provides rationale for clinical trials of RT combined with defined ICI therapy, and these findings will support future studies to search for more effective targets and timing of therapy administration.
Collapse
Affiliation(s)
- Shunsuke A. Sakai
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Koichi Saeki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - SungGi Chi
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Yamato Hamaya
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Junyan Du
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Masaki Nakamura
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Hidehiro Hojo
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Takashi Kojima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hideaki Bando
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Motohiro Kojima
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
- Pathology Division, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Ayako Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Tetsuo Akimoto
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Katsuya Tsuchihara
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Hiroshi Haeno
- Division of Integrated Research, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Riu Yamashita
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Shun-Ichiro Kageyama
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| |
Collapse
|
43
|
Fuereder T, Kocher F, Vermorken JB. Systemic therapy for laryngeal carcinoma. Front Oncol 2025; 15:1541385. [PMID: 40104492 PMCID: PMC11913863 DOI: 10.3389/fonc.2025.1541385] [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: 12/07/2024] [Accepted: 02/07/2025] [Indexed: 03/20/2025] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) accounts for 100,000 deaths worldwide each year. Despite multimodal treatment, outcomes for both high-risk locally advanced and recurrent/metastatic laryngeal carcinoma remain poor. Treatment intensification through induction chemotherapy has not improved overall survival, although it may contribute to larynx preservation. Consequently, multiple recent efforts have been made to integrate novel immunotherapies into the current treatment algorithm for LSCC. In particular, perioperative immunotherapy regimens appear to be the most promising approach for preserving laryngeal function and optimizing event-free and overall survival rates in the locally advanced setting. In the recurrent/metastatic setting, the 5-year overall survival rate is approximately 20% with pembrolizumab-based regimens. Primary and secondary resistance to immunotherapy is frequently observed in the majority of patients. Along with trials of checkpoint inhibitor monotherapy, combinatorial approaches with novel immunotherapies, bispecific antibodies, targeted therapies, and antibody-drug conjugates are being explored for the treatment of recurrent/metastatic laryngeal carcinoma. This article aims to discuss recent efforts to improve outcomes and quality of life for patients with locally advanced and recurrent/metastatic LSCC.
Collapse
Affiliation(s)
- Thorsten Fuereder
- Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Florian Kocher
- Department of Internal Medicine V (Hematology and Oncology), Medical University of Innsbruck, Innsbruck, Austria
| | - Jan Baptist Vermorken
- Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Department of Medical Oncology, Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|
44
|
Courtney PT, Venkat PS, Shih YCT, Chang AJ, Lee A, Steinberg ML, Raldow AC. Cost-Effectiveness of Pembrolizumab With Chemoradiotherapy for Locally Advanced Cervical Cancer. JAMA Netw Open 2025; 8:e250033. [PMID: 40036034 PMCID: PMC11880949 DOI: 10.1001/jamanetworkopen.2025.0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 12/19/2024] [Indexed: 03/06/2025] Open
Abstract
Importance The KEYNOTE-A18 trial demonstrated that adding concurrent and adjuvant pembrolizumab to chemoradiotherapy and brachytherapy significantly improved survival in patients with newly diagnosed, locally advanced cervical cancer. However, considering the annual global incidence of 660 000 cases of cervical cancer, including 13 820 in the US in 2024, incorporating this regimen into the standard of care could have substantial health care economic implications for both patients and the health care system. Objective To determine the cost-effectiveness of adding pembrolizumab to the first-line treatment of newly diagnosed, locally advanced cervical cancer. Design, Setting, and Participants This economic evaluation created a Markov model simulating 50-year outcomes to evaluate cost-effectiveness from the payer perspective for patients receiving either pembrolizumab or placebo in addition to chemoradiotherapy plus brachytherapy. Probabilities, including disease progression, survival, and treatment-related toxic effects, were derived from KEYNOTE-A18 clinical trial data in patients with newly diagnosed, locally advanced cervical cancer. Costs and health utilities were obtained from published literature; 1-way, 3-way, and probabilistic sensitivity analyses were used to assess model uncertainty. Data analyses were conducted from April to November 2024. Exposure Pembrolizumab. Main Outcomes and Measures Costs, measured in 2024 US dollars, and effectiveness, measured in quality-adjusted life-years (QALYs) were used to calculate an incremental cost-effectiveness ratio (ICER). A willingness-to-pay threshold of $100 000 per QALY was chosen, below which pembrolizumab would be considered cost-effective. Results KEYNOTE-A18 enrolled 1060 patients (529 in pembrolizumab group, 531 in placebo group). The median age was 50 years. Pembrolizumab increased costs by $257 000 and effectiveness by 1.40 QALYs, yielding an incremental cost-effectiveness ratio of $183 400 per QALY. The addition of pembrolizumab became cost-effective if its monthly cost was decreased from $16 990 to $9190 (a 45.6% reduction) or its maximum duration of 24 months was decreased to 10 months. The model was insensitive to assumptions about treatment-related toxic effects, progression-free survival, and overall survival. Probabilistic sensitivity analysis indicated that at a willingness-to-pay threshold of $100 000 per QALY, the addition of pembrolizumab was cost-effective 37.3% of the time. Conclusions and Relevance In this economic evaluation of adding concurrent and adjuvant pembrolizumab to first-line treatment of newly diagnosed, locally advanced cervical cancer, this regimen was not cost-effective at current prices despite data demonstrating improved survival with this regimen.
Collapse
Affiliation(s)
| | - Puja S. Venkat
- Department of Radiation Oncology, University of California, Los Angeles
| | - Ya-Chen Tina Shih
- Department of Radiation Oncology, University of California, Los Angeles
| | - Albert J. Chang
- Department of Radiation Oncology, University of California, Los Angeles
| | - Alan Lee
- Department of Radiation Oncology, University of California, Los Angeles
| | | | - Ann C. Raldow
- Department of Radiation Oncology, University of California, Los Angeles
| |
Collapse
|
45
|
Liu Y, Yang Z, Pu JJ, Zhong J, Khoo U, Su Y, Zhang G. Proteogenomic characterisation of primary oral cancer unveils extracellular matrix remodelling and immunosuppressive microenvironment linked to lymph node metastasis. Clin Transl Med 2025; 15:e70261. [PMID: 40038875 PMCID: PMC11879901 DOI: 10.1002/ctm2.70261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/08/2025] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC) is an increasingly prevalent malignancy worldwide. This study aims to understand molecular alterations associated with lymph node metastasis of OSCC in order to improve treatment strategies. We analysed a cohort of 46 patients with primary OSCC, including 10 with lymph node metastasis and 36 without. Using a comprehensive multi-omics approach - encompassing genomic, transcriptomic, proteomic, epigenetic, single-cell, and spatial analyses - we integrated data to delineate the molecular landscape of OSCC in the context of lymph node metastasis. Our genomic analysis identified significant mutations in key genes within the MAPK, TGF-β and WNT signalling pathways, which are essential for tumour development. The proteogenomic analysis highlighted pathways critical for lymph node dissemination and factors contributing to an immunosuppressive tumour microenvironment. Elevated levels of POSTN were found to reorganise the extracellular matrix (ECM), interact with TGF-β, disrupt cell cycle regulation and suppress the immune response by reducing VCAM1 activity. Integrated analyses of single-cell and spatial transcriptome data revealed that cancer-associated fibroblasts (CAFs) secrete TGF-β1/2, promoting cancer cell metastasis through epithelial-mesenchymal transition (EMT). Our integrated multi-omics analysis provides a detailed understanding of molecular mechanisms driving lymph node metastasis of OSCC. These insights could lead to more precise diagnostics and targeted treatments. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yu Liu
- Department of Thoracic Surgery/Institute of Thoracic OncologyWest China HospitalSichuan UniversityChengduChina
- Faculty of DentistryThe University of Hong KongHong KongHong Kong
| | - Zhenyu Yang
- Department of Thoracic Surgery/Institute of Thoracic OncologyWest China HospitalSichuan UniversityChengduChina
| | - Jingya Jane Pu
- Faculty of DentistryThe University of Hong KongHong KongHong Kong
| | - Jie Zhong
- Faculty of DentistryThe University of Hong KongHong KongHong Kong
| | - Ui‐Soon Khoo
- Department of PathologySchool of Clinical MedicineThe University of Hong KongHong KongHong Kong
| | - Yu‐Xiong Su
- Faculty of DentistryThe University of Hong KongHong KongHong Kong
| | - Gao Zhang
- Faculty of DentistryThe University of Hong KongHong KongHong Kong
| |
Collapse
|
46
|
Dong S, Li X, Huang Q, Li Y, Li J, Zhu X, Xue C, Chen R, Zeng Y, Wu J, Zhong Y, Hu S. Resistance to immunotherapy in non-small cell lung cancer: Unraveling causes, developing effective strategies, and exploring potential breakthroughs. Drug Resist Updat 2025; 81:101215. [PMID: 40081220 DOI: 10.1016/j.drup.2025.101215] [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: 08/21/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 03/15/2025]
Abstract
Over the last two decades, advancements in deciphering the intricate interactions between oncology and immunity have fueled a meteoric rise in immunotherapy for non-small cell lung cancer, typified by an explosive growth of immune checkpoint inhibitors. However, resistance to immunotherapy remains inevitable. Herein we unravel the labyrinthine mechanisms of resistance to immunotherapy, characterized by their involvement of nearly all types of cells within the body, beyond the extrinsic cancer cells, and importantly, such cells are not only (inhibitory or excitatory, or both) signal recipients but also producers, acting in a context-dependent manner. At the molecular level, these mechanisms underlie genetic and epigenetic aberrations, which are regulated by or regulate various protein kinases, growth factors, and cytokines with inherently dynamic and spatially heterogeneous properties. Additionally, macroscopic factors such as nutrition, comorbidities, and the microbiome within and around organs or tumor cells are involved. Therefore, developing therapeutic strategies combined with distinct action informed by preclinical, clinical, and real-world evidence, such as radiotherapy, chemotherapy, targeted therapy, antibody-drug conjugates, oncolytic viruses, and cell-based therapies, may stand as a judicious reality, although the ideality is to overcome resistance point-by-point through a novel drug. Notably, we highlight a realignment of treatment aims, moving the primary focus from eliminating cancer cells -- such as through chemotherapy and radiotherapy -- to promoting immune modulation and underscore the value of regulating various components within the host macro- or micro-environment, as their effects, even if seemingly minimal, can cumulatively contribute to visible clinical benefit when applied in combination with ICIs. Lastly, this review also emphasizes the current hurdles scattered throughout preclinical and clinical studies, and explores evolving directions in the landscape of immunotherapy for NSCLC.
Collapse
Affiliation(s)
- Shuang Dong
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Xiaoyu Li
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Qing Huang
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Yuanxiang Li
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | | | - Xianmin Zhu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Chang Xue
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Runzhi Chen
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Yuan Zeng
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Jingyi Wu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Yi Zhong
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China.
| | - Sheng Hu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China.
| |
Collapse
|
47
|
Zschaeck S, Hajiyianni M, Hausmann P, Nikulin P, Kukuk E, Furth C, Cegla P, Lombardo E, Kazmierska J, Holzgreve A, Strouthos I, Stromberger C, Belka C, Baumann M, Krause M, Landry G, Cholewinski W, Kotzerke J, Zips D, van den Hoff J, Hofheinz F. Total lesion glycolysis of primary tumor and lymphnodes is a strong predictor for development of distant metastases in oropharyngeal carcinoma patients with independent validation in automatically delineated lesions. Cancer Imaging 2025; 25:18. [PMID: 39985091 PMCID: PMC11844016 DOI: 10.1186/s40644-025-00836-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 02/06/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Oropharyngeal carcinomas are characterized by an increasing incidence and a relatively good prognosis. Nonetheless, a considerable number of patients develops metachronous distant metastases; identification of these patients is an urgent medical need. METHODS This is a retrospective multicenter evaluation of 431 patients. All patients underwent [18F]-FDG positron emission tomography (PET). The cohort was split into an explorative group (n = 366) and a validation group (n = 65). Lesions were manually delineated in the explorative group and automatically delineated by a convolutional neuronal network (CNN) in the validation group. Quantitative PET parameters standardized uptake value (SUV), metabolic tumor volume (MTV), and total lesion glycolysis (TLG) were calculated for primary tumors (prim) and tumor plus lymphnodes (all). Association of parameters with freedom from distant metastases (FFDM) and overall survival (OS) was tested by cox regression analyses. RESULTS In the explorative group, univariate analyses revealed an association of metric MTVprim (p = 0.022), MTVall (p < 0.001) and TLGall (p < 0.001) with FFDM, binarized parameters were also associated with FFDM (p < 0.001 and p = 0.002). Bootstrap analyses revealed a significantly better association of TLGall compared to TLGprim with FFDM (p = 0.02). MTVall and TLGall remained significantly associated with FFDM upon multivariate testing (p = 0.002, p = 0.031, respectively). In the validation group, the cutoff value for TLGall but not for TLGprim was significantly associated with FFDM (HR = 3.1, p = 0.045). Additional analyses with manually delineated contours of the validation cohort revealed a similar effect (HR = 3.47, p = 0.026). No considerable differences between HPV positive and negative disease were observed. CONCLUSIONS TLGall is a promising biomarker to select OPC patients with high risk for metachronous distant metastases.
Collapse
Affiliation(s)
- Sebastian Zschaeck
- Department of Radiation Oncology, Charité- Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany.
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité (Junior) Clinician Scientist Program, Berlin, Germany.
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- German Cancer Consortium (DKTK), partner site Dresden, Germany, and German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany.
- OncoRay- National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden- Rossendorf, Dresden, Germany.
| | - Marina Hajiyianni
- Department of Radiation Oncology, Charité- Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Patrick Hausmann
- Department of Radiation Oncology, Charité- Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Pavel Nikulin
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Emily Kukuk
- Department of Radiation Oncology, Charité- Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Christian Furth
- Department of Nuclear Medicine, Charité Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Paulina Cegla
- Department of Nuclear Medicine, Greater Poland Cancer Centre, Poznan, Poland
| | - Elia Lombardo
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Joanna Kazmierska
- Electroradiology Department, University of Medical Sciences, Poznan, Poland
- Radiotherapy Department II, Greater Poland Cancer Centre, Poznan, Poland
| | - Adrien Holzgreve
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Iosif Strouthos
- Department of Radiation Oncology, German Oncology Center, European University Cyprus, Limassol, Cyprus
| | - Carmen Stromberger
- Department of Radiation Oncology, Charité- Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Claus Belka
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU University Hospital Munich, Munich, Germany
| | - Michael Baumann
- OncoRay- National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden- Rossendorf, Dresden, Germany
- Division of Radiooncology/Radiobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mechthild Krause
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), partner site Dresden, Germany, and German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany
- OncoRay- National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden- Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU University Hospital Munich, Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- Institute of Radiooncology, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Guillaume Landry
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Witold Cholewinski
- Department of Nuclear Medicine, Greater Poland Cancer Centre, Poznan, Poland
- Electroradiology Department, University of Medical Sciences, Poznan, Poland
| | - Jorg Kotzerke
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Daniel Zips
- Department of Radiation Oncology, Charité- Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité- Universitätsmedizin Berlin, Berlin, Germany
- National Tumor Center Berlin (NCT), Partner Site Berlin, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany
- Charité Comprehensive Cancer, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, Charité Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Centrum für Molekulare Medizin, Helmholtz Association, Berlin, Germany
| | - Jörg van den Hoff
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Frank Hofheinz
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| |
Collapse
|
48
|
Kanai M. Challenges and Opportunities for the Clinical Application of the Combination of Immune-Checkpoint Inhibitors and Radiation Therapy in the Treatment of Advanced Pancreatic Cancer. Cancers (Basel) 2025; 17:606. [PMID: 40002201 PMCID: PMC11853451 DOI: 10.3390/cancers17040606] [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: 01/05/2025] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
The treatment landscape of pancreatic ductal adenocarcinoma (PDAC) has seen slow progress, with immune-checkpoint inhibitors (ICIs) failing to replicate the success observed in other malignancies. The immune-suppressive tumor microenvironment (TME) in PDAC represents a significant barrier, limiting the activation of an effective antitumor immune response following ICI administration. Radiation therapy (RT), with its immunomodulatory effects, has emerged as a promising partner for ICIs. This review discusses the recent efforts evaluating the combination of ICIs and RT in advanced PDAC. While the combination therapy has demonstrated an acceptable safety profile, the reported clinical efficacy remains modest, particularly for patients with refractory metastatic PDAC. The ongoing phase III trial (JCOG1908E) will clarify whether the combination of ICI and RT improves overall survival in chemo-naïve patients with locally advanced PDAC.
Collapse
Affiliation(s)
- Masashi Kanai
- Department of Clinical Oncology, Kansai Medical University Hospital, 3-1 Shinmachi 2 Chome, Hirakata City 573-1191, Osaka, Japan
| |
Collapse
|
49
|
Filippini DM, Broseghini E, Liberale C, Gallerani G, Siepe G, Nobili E, Ferracin M, Molteni G. Vaccine-Based Immunotherapy for Oropharyngeal and Nasopharyngeal Cancers. J Clin Med 2025; 14:1170. [PMID: 40004705 PMCID: PMC11856027 DOI: 10.3390/jcm14041170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/05/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Viral infections such as human papillomavirus (HPV) and Epstein-Barr virus (EBV) play a critical role in the onset of oropharyngeal (OPC) and nasopharyngeal cancer (NPC), respectively. Despite advancements in targeted therapies and immunotherapies, in the recurrent/metastatic setting, these tumors remain incurable diseases with poor prognosis. The development of therapeutic tumor vaccines, utilizing either neoantigens or oncoviral antigens, represents a promising addition to the cancer immunotherapy arsenal. Research on vaccine-based immunotherapy for OPC and NPC focuses on targeting viral antigens, particularly HPV E6/E7 and EBV EBNA1/LMP2. The potential for vaccine platforms, including peptide-based, DNA, RNA, and viral vector-based vaccines, to induce durable immune responses against viral antigens is reported. The early-phase clinical trials evaluating vaccine-based therapies for HPV-related OPC and EBV-related NPC revealed safety and preliminary signs of efficacy; however, further clinical trials are crucial for validation. This review provides an overview of the current landscape of vaccine-based strategies for HPV-related OPC and EBV-related NPC, discussing their biological mechanisms and immune processes involved in anti-HPV and anti-EBV vaccine treatments, with a particular focus on the immune factors that influence these therapies.
Collapse
Affiliation(s)
- Daria Maria Filippini
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy; (G.G.); (M.F.)
| | | | - Carlotta Liberale
- Unit of Otorhinolaryngology, Head & Neck Department, University of Verona, 37134 Verona, Italy;
| | - Giulia Gallerani
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy; (G.G.); (M.F.)
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Giambattista Siepe
- Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Elisabetta Nobili
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Manuela Ferracin
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy; (G.G.); (M.F.)
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Gabriele Molteni
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy; (G.G.); (M.F.)
- Department of Otolaryngology-Head and Neck Surgery, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
50
|
Liang H, Liang W, Tian J, Zheng S, Su Y, Huang P, Chen R, Guan Z, Cai Q. Safety and efficacy of neoadjuvant therapy with tislelizumab plus chemotherapy for locally advanced head and neck squamous cell carcinoma: a single-arm, retrospective study. Cancer Immunol Immunother 2025; 74:108. [PMID: 39932534 PMCID: PMC11813832 DOI: 10.1007/s00262-025-03953-0] [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: 11/07/2024] [Accepted: 01/21/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is a very common type of head and neck cancer, and the 5-year overall survival (OS) rate is only 50%. Inhibitors targeting the programmed death 1 (PD-1) pathway have gained widespread clinical adoption. However, the relationship between the infiltration characteristics of immune cells in the tumor immune microenvironment (TIME) and the effect of immunotherapy on HNSCC remains to be explored. METHODS Patients diagnosed with HNSCC who received Tislelizumab combined with chemotherapy were reviewed between February 2021 and March 30, 2024, in our single center. The laryngoscopy, magnetic resonance imaging (MRI), and pathologic response were evaluated to the efficacy of neoadjuvant therapy with Tislelizumab plus chemotherapy treatment. Treatment-related adverse events (TRAEs) were evaluated according to the Common Terminology Criteria for Adverse Events version 5.0. RESULT Our analysis involved 42 patients who received Tislelizumab combined with chemotherapy. A total of 18 patients underwent surgical treatment following the completion of immunotherapy combined with chemotherapy. Among them, 6 patients achieved pCR (33%). The 1-year OS rate of the 42 patients enrolled in the study was 95.05%, and the 1-year PFS rate was 89.86%. There was a significant positive correlation between the lymphocyte density in HNSCC prior to the administration of neoadjuvant PD-1 inhibitor therapy combined with chemotherapy and the immunotherapy efficacy. Compared with pretreatment, the neutrophil-to-lymphocyte ratio (NLR) was significantly decreased, and the lymphocyte density was significantly increased in HNSCC patients after immunotherapy. CONCLUSIONS The integration of neoadjuvant PD-1 inhibitor therapy with chemotherapy has been demonstrated to be a safe and effective strategy, can improve the tumor response rate and survival rate, and is a valuable treatment for patients with HNSCC. Furthermore, the study suggests that an elevated NLR within the HNSCC tumor microenvironment could potentially serve as a biomarker indicative of diminished immunotherapy efficacy.
Collapse
Affiliation(s)
- Haifeng Liang
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China
| | - Wenting Liang
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China
| | - Jiawang Tian
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China
| | - Shibei Zheng
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China
- Department of Otorhinolaryngology, Head and Neck Surgery, Shenzhen Baoan District People's Hospital, Shenzhen, China
| | - Yangzhou Su
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China
| | - Piao Huang
- Department of Pathology, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China
| | - Renhui Chen
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China.
| | - Zhong Guan
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China.
| | - Qian Cai
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China.
- Department of Otolaryngology-Head and Neck, Zhujiang Hospital of Southern Medical University, Guangzhou, China.
| |
Collapse
|