51
|
Vanpouille-Box C. Recent Mechanistic Insight into the Immunogenic Properties of Radiation-Induced Micronuclei. Int J Radiat Oncol Biol Phys 2025; 121:283-286. [PMID: 39824567 DOI: 10.1016/j.ijrobp.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 11/03/2024] [Indexed: 01/20/2025]
Affiliation(s)
- Claire Vanpouille-Box
- Department of Radiation Oncology, Weill Cornell Medicine, New York; Sandra and Edward Meyer Cancer Center, New York.
| |
Collapse
|
52
|
Zhao M, Schoenfeld JD, Egloff AM, Hanna GJ, Haddad RI, Adkins DR, Uppaluri R. T cell dynamics with neoadjuvant immunotherapy in head and neck cancer. Nat Rev Clin Oncol 2025; 22:83-94. [PMID: 39658611 DOI: 10.1038/s41571-024-00969-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2024] [Indexed: 12/12/2024]
Abstract
Immune-checkpoint inhibitors (ICIs) are being tested as neoadjuvant therapies in various solid tumours, including in patients with head and neck squamous cell carcinoma (HNSCC), with promising results. Key findings thus far include that this approach is well-tolerated with favourable clinical outcomes including promising pathological response rates in initial studies. Pathological responses are likely to be increased by combining other agents with anti-PD-(L)1 antibodies. Comparisons of baseline biopsy samples with post-treatment surgical specimens have enabled correlative studies utilizing multiomic and immunogenomic methods. Data from these studies suggest that pretreatment intratumoural tissue-resident memory CD8+ T cells are key drivers of tumour regression and give rise to both local and systemic antitumour immune responses. Analyses of systemic responses have defined a PD-1+KLRG1- circulating CD8+ T cell subpopulation that is highly predictive of response, and revealed the interrelationships between intratumoural clones and circulating CD8+ T cells. Lastly, interrogation of T cell populations within lymph nodes is beginning to delineate the immune crosstalk between the primary tumour and tumour-draining lymph nodes and how this relationship might be disrupted with tumour infiltration of the latter. In this Review, we examine data from trials testing neoadjuvant ICIs in patients with HNSCC, focusing on human papillomavirus-unrelated disease, and highlight correlative immunogenomic findings from these trials.
Collapse
Affiliation(s)
- Maryann Zhao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jonathan D Schoenfeld
- Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Boston, MA, USA
| | - Ann Marie Egloff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Glenn J Hanna
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Robert I Haddad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Douglas R Adkins
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine/Medical Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ravindra Uppaluri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
53
|
Clark P, Karasik N, Campbell SR, Woody NM, Ku JA, Silver N, Bottalico D, Prendes BL, Lamarre ED, Scharpf J, Sussman TA, Geiger JL, Wang H, Chan TA, Koyfman SA, Miller JA. Highly-multiplex detection of plasma cell-free human papillomavirus-16 DNA in oropharyngeal carcinoma. J Clin Virol 2025; 176:105760. [PMID: 39793474 DOI: 10.1016/j.jcv.2025.105760] [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] [Revised: 12/30/2024] [Accepted: 01/01/2025] [Indexed: 01/13/2025]
Abstract
BACKGROUND Plasma cell-free Human Papillomavirus DNA (cfHPVDNA) is a biomarker for oropharyngeal carcinoma. Existing diagnostics may be limited by inadequate sensitivity or high cost/complexity for longitudinal monitoring. OBJECTIVES We hypothesized that sensitive and specific plasma cfHPVDNA detection may be achieved via a highly-multiplex qPCR method. STUDY DESIGN We designed and validated a single-tube one-step genotype-specific qPCR assay for detection of cfHPV16DNA in human plasma using >8,000 genomes spanning 18 genotypes. Amplicons were optimized for cfHPVDNA fragment size. RESULTS The cfHPV16DNA qPCR amplicons spanned 16 % of the HPV16 genome. Amplicons were conserved in a median of 99.0 % of 3,944 genomes in silico. The 95 % lower limit of detection was 0.35 genome copies/reaction and the limit of blank was 0. Multiplexing achieved a tenfold improvement in sensitivity compared with single amplicons using in silico simulations of cfHPVDNA fragmentation, which was in close agreement with experimental observations. An assay was replicated for HPV18 with similar observations. Among 36 patients with head/neck mucosal carcinomas (26 HPV-positive, 12 HPV-negative), there was 100 % concordance with tissue HPV status and with NavDx digital PCR. Pre-treatment specimens with sub-genomic cfHPVDNA concentration were detected. False negatives were observed with single amplicons but not with this multiplexed method. Among 17 patients with post-treatment landmark specimens, there was 100 % PPV and 100 % NPV for recurrence. CONCLUSIONS This assay is specific for plasma cfHPVDNA detection and prognostic for recurrence. Sub-genomic sensitivity was in close agreement with in silico simulations. The format might be more accessible than dPCR or NGS for longitudinal testing.
Collapse
Affiliation(s)
- Payton Clark
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Natalya Karasik
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Shauna R Campbell
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Neil M Woody
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jamie A Ku
- Department of Head and Neck Surgery, Head and Neck Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Natalie Silver
- Department of Head and Neck Surgery, Head and Neck Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Danielle Bottalico
- Department of Head and Neck Surgery, Head and Neck Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Brandon L Prendes
- Department of Head and Neck Surgery, Head and Neck Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Eric D Lamarre
- Department of Head and Neck Surgery, Head and Neck Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Joseph Scharpf
- Department of Head and Neck Surgery, Head and Neck Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tamara A Sussman
- Department of Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jessica L Geiger
- Department of Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Hannah Wang
- Department of Laboratory Medicine, Diagnostics Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Timothy A Chan
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Shlomo A Koyfman
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jacob A Miller
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
54
|
Lin H, Heng Y, Zhu X, Wang T, Tao L. Impact of radiotherapy on peripheral T lymphocytes in hypopharyngeal squamous cell carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol 2025; 139:220-228. [PMID: 39523138 DOI: 10.1016/j.oooo.2024.10.080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/05/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND To investigate the impact of radiotherapy (RT) on the distribution and function of peripheral CD8+ T lymphocytes in patients with hypopharyngeal squamous cell carcinoma (HPSCC). METHODS A total of 105 HPSCC patients who underwent definitive RT were enrolled. Baseline levels of peripheral immune cells were obtained, and their alteration during RT was evaluated. Flow cytometry was used to analyze T-cell distribution, cytokine secretion, and CD8+ T lymphocyte proliferation capacity. RESULTS Lymphocyte count significantly decreased following radiation and remained in a low level after 1 year of RT. CD3+ T lymphocyte counts decreased significantly, and the CD4+/CD8+ ratio increased in HPSCC patients following radiation. The secretion of IFN-γ from peripheral CD8+ T lymphocytes was significantly reduced after irradiation, while the secretion of TNF-α and perforin did not change significantly. Furthermore, the proliferation capacity of peripheral CD8+ T lymphocytes was decreased following RT. CONCLUSIONS RT significantly decreased the number of peripheral T lymphocytes and impaired the secretory function and proliferation ability of CD8+ T lymphocytes in HPSCC patients. These findings provide insight into the mechanisms underlying the therapeutic effects of RT on HPSCC and have implications for optimizing treatment strategies.
Collapse
Affiliation(s)
- Hanqing Lin
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, PR China; Department of Otolaryngology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China
| | - Yu Heng
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, PR China
| | - Xiaoke Zhu
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, PR China
| | - Tian Wang
- Department of Radiation Oncology, Eye & ENT Hospital, Fudan University, Shanghai, PR China
| | - Lei Tao
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, PR China.
| |
Collapse
|
55
|
Zhang YN, Chen YP, OuYang PY, Lu TX, Xie FY, Han F, Chen CY. Anti-programmed death-1 inhibitors and nimotuzumab in combination with induction chemotherapy for locoregionally advanced nasopharyngeal carcinoma: a propensity score-matched analysis. Ther Adv Med Oncol 2025; 17:17588359251316094. [PMID: 39896749 PMCID: PMC11783488 DOI: 10.1177/17588359251316094] [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: 10/08/2024] [Accepted: 01/09/2025] [Indexed: 02/04/2025] Open
Abstract
Background The poor prognosis of locoregionally advanced nasopharyngeal carcinoma (LANPC) due to the high incidence of metastasis necessitates effective treatment strategies. Synergistic effects have been observed when anti-programmed death-1 (PD-1) inhibitors are combined with chemotherapy or targeted therapy. Objectives To compare the efficacy and safety of induction chemotherapy in combination with nimotuzumab with or without anti-PD-1 inhibitors for LANPC. Design Retrospective study. Methods In total, 319 patients with LANPC were retrospectively enrolled between December 2017 and November 2022. The primary endpoint was progression-free survival (PFS). Propensity score matching was performed to adjust for potential confounders. Results Overall, 150 patients were included after propensity score matching. The immunotherapy + nimotuzumab + chemotherapy (INC) group (n = 50) had a higher 3-year PFS rate (96.6% (95% confidence interval (CI): 93.2-100.0)) than the nimotuzumab + chemotherapy (NC) group (n = 100) (79.8% (95% CI: 75.6-84.0)). The INC group had a hazard ratio of 0.16 (95% CI: 0.02-1.22; p = 0.04). The objective response rates were 100% and 99% for the INC and NC groups, respectively. Grade ⩾3 treatment-related adverse events were reported in eight (5.3%) patients, and hyponatremia (2.0%) was the most common. Grade ⩾3 immune-related adverse events (rash and reactive capillary proliferation) were reported in two (4.0%) patients. Conclusion INC demonstrated remarkable anti-tumor activity with acceptable safety for LANPC.
Collapse
Affiliation(s)
- Ya-Ni 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, Guangdong, 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, Guangdong, China
| | - Pu-Yun OuYang
- 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, China
| | - Tai-Xiang 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, Guangdong, China
| | - Fang-Yun Xie
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong 510060, China
| | - Fei Han
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong 510060, China
| | - Chun-Yan Chen
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong 510060, China
| |
Collapse
|
56
|
Jiang C, Wang S, Zhu L. Efficacy and safety of immunotherapy for head and neck squamous cell carcinoma: a meta-analysis of randomized clinical trials. Front Oncol 2025; 14:1489451. [PMID: 39850817 PMCID: PMC11755100 DOI: 10.3389/fonc.2024.1489451] [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: 09/01/2024] [Accepted: 12/09/2024] [Indexed: 01/25/2025] Open
Abstract
Background Head and neck squamous cell carcinoma (HNSCC) is one of the most common types of cancer worldwide and immune checkpoint inhibitors have shown favorable therapeutic effects in recurrent or metastatic or locally advanced head and neck squamous cell carcinoma (R/M/LA HNSCC). However, the effects of immunotherapy in HNSCC are still inconsistent because of complicating factors. This meta-analysis tries to provide a more precise assessment of the efficacy and safety of this integrated approach in HNSCC. Methods We conducted a systematic review and meta-analysis of randomized clinical trials according to Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) reporting guidelines. The outcomes were overall survival (OS), progression-free survival (PFS), and treatment-related adverse events (TRAEs). A total of 8 out of 2445 articles were analyzed, including 5067 HNSCC patients, including 823 and 4244 patients with LA HNSCC and R/M HNSCC. Results The combined data revealed that immunotherapy has an apparent difference in OS (HR 0.86 95% CI 0.77-0.98) compared with standard of care (Soc, like fluoropyrimidine, methotrexate, docetaxel, or cetuximab) but was equal with the other treatment in PFS (HR 1.08, 95% CI 0.85-1.37). Furthermore, the occurrence of grade 3 or higher adverse events related to the drugs was lower than systematic therapy (OR 0.35, 95% CI 0.17-0.73). Conclusions The study has provided compelling evidence that immunotherapy is a significant benefit in OS for HNSCC patients, either R/M HNSCC or LA HNSCC, immunochemotherapy may benefit more for these patients, but double-agent immunotherapy showed no more benefit for R/M HNSCC patients. Systematic Review Registration https://www.crd.york.ac.uk/, identifier CRD42023471570.
Collapse
Affiliation(s)
| | | | - Lijun Zhu
- Department of Oral and Maxillofacial Surgery, Guangdong Provincial People’s
Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
57
|
Spindler KLG, Jakobsen AV, Eriksen JG, Fokdal L, Nordsmark M, Thorsen LBJ, Wind KL, Lefevre AC, Overgaard J. The clinical utility of circulating human papillomavirus across squamous cell carcinomas. Acta Oncol 2025; 64:1-12. [PMID: 39748655 PMCID: PMC11711493 DOI: 10.2340/1651-226x.2025.41288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/08/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND AND PURPOSE The similarities in biology, treatment regimens and outcome between the different human papillomavirus (HPV) associated squamous cell carcinomas (SCCs) allow for extrapolation of results generated from one SC tumor type to another. In HPV associated cancers, HPV is integrated into the tumor genome and can consequently be detected in the circulating fragments of the tumor DNA. Thus, measurement of HPV in the plasma is a surrogate for circulating tumor DNA (ctDNA) and holds promise as a clinically relevant biomarker in HPV associated cancers. With the present overview we aim to present the status of circulating HPV studies in SCCs, the clinical potential and the gaps of knowledge, with the overall aim to facilitate the next steps into clinically relevant prospective trials. MATERIAL AND METHODS We reviewed the literature and presented the data for each tumor type as well as analyses of the clinical utility across the SCC. RESULTS AND INTERPRETATION A total of 41 studies were identified in cervical, head and neck and anal SCC and we discuss the common signals from the results across the different tumor sites. Our results not only confirm the strong clinical potential but also emphasize an urgent need to coordinate studies to allow for relevant sample sizes and statistical validations.
Collapse
Affiliation(s)
- Karen-Lise G Spindler
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Anne V Jakobsen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jesper G Eriksen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Lars Fokdal
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Oncology, Vejle Hospital, Vejle, Denmark
| | - Marianne Nordsmark
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Lise B J Thorsen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Karen L Wind
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anna C Lefevre
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Overgaard
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
58
|
Chen Y, Yang X, Li Q. PDLIM1 Inhibits Chemoresistance by Blocking DNA Damage Repair in Gastric Cancer. Recent Pat Anticancer Drug Discov 2025; 20:260-273. [PMID: 38779728 DOI: 10.2174/0115748928307544240502064448] [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: 02/21/2024] [Revised: 04/06/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE Current cisplatin (CDDP) resistance remains a major challenge in the treatment of advanced gastric cancer. To address the issue of drug resistance, we explored the regulatory functions of PDZ and LIM structural domain protein 1 (PDLIM1) in CDDP chemotherapy for gastric cancer. METHODS In this study, we analyzed PDLIM1 expression and prognosis using bioinformatics on publicly available data. PDLIM1 expression in a gastric mucosal epithelial cell line (GSE-1), CDDP- sensitive (SGC7901, BGC823) and CDDP-resistant gastric cancer cells was detected by RTqPCR and Western blotting. Cell proliferative capacity was assessed by knockdown of PDLIM1 and overexpression of PDLIM1 in cells administered in combination with cisplatin, and apoptotic levels were measured by CCK-8 and colony formation assay and by flow cytometry. Expression of breast cancer susceptibility gene 1 (BRCA1) and γH2AX was determined by Western blotting or immunofluorescence staining. RESULTS Downregulation of PDLIM1 was found in tumor tissues and cells, which was associated with poor clinical outcomes. Knockdown of PDLIM1 enhanced proliferation and attenuated apoptosis in gastric cancer cells. In addition, the therapeutic effects of CDDP on proliferation, apoptosis, and DNA damage repair were attenuated by PDLIM1 deletion.PDLIM1 expression was downregulated in CDDP-resistant tumor cells. Overexpression of PDLIM1 overcomes CDDP resistance in tumor cells as BRCA1 expression decreases and γH2AX expression increases. CONCLUSION Our findings demonstrate that PDLIM1 enables to alleviate gastric cancer progression and resistance to cisplatin via impeding DNA damage repair.
Collapse
Affiliation(s)
- Yuli Chen
- Health Management Center, The Third People's Hospital of Chengdu, Sichuan, Chengdu, 610014, China
| | - Xin Yang
- School of Clinical Medicine, Chengdu Medical College, Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Qiang Li
- Department of Emergency, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| |
Collapse
|
59
|
Catanzaro E, Beltrán-Visiedo M, Galluzzi L, Krysko DV. Immunogenicity of cell death and cancer immunotherapy with immune checkpoint inhibitors. Cell Mol Immunol 2025; 22:24-39. [PMID: 39653769 PMCID: PMC11685666 DOI: 10.1038/s41423-024-01245-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024] Open
Abstract
While immunotherapy with immune checkpoint inhibitors (ICIs) has revolutionized the clinical management of various malignancies, a large fraction of patients are refractory to ICIs employed as standalone therapeutics, necessitating the development of combinatorial treatment strategies. Immunogenic cell death (ICD) inducers have attracted considerable interest as combinatorial partners for ICIs, at least in part owing to their ability to initiate a tumor-targeting adaptive immune response. However, compared with either approach alone, combinatorial regimens involving ICD inducers and ICIs have not always shown superior clinical activity. Here, we discuss accumulating evidence on the therapeutic interactions between ICD inducers and immunotherapy with ICIs in oncological settings, identify key factors that may explain discrepancies between preclinical and clinical findings, and propose strategies that address existing challenges to increase the efficacy of these combinations in patients with cancer.
Collapse
Affiliation(s)
- Elena Catanzaro
- Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Manuel Beltrán-Visiedo
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| | - Dmitri V Krysko
- Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.
| |
Collapse
|
60
|
de Bakker T, Maes A, Dragan T, Martinive P, Penninckx S, Van Gestel D. Strategies to Overcome Intrinsic and Acquired Resistance to Chemoradiotherapy in Head and Neck Cancer. Cells 2024; 14:18. [PMID: 39791719 PMCID: PMC11719474 DOI: 10.3390/cells14010018] [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: 11/19/2024] [Revised: 12/18/2024] [Accepted: 12/25/2024] [Indexed: 01/12/2025] Open
Abstract
Definitive chemoradiotherapy (CRT) is a cornerstone of treatment for locoregionally advanced head and neck cancer (HNC). Research is ongoing on how to improve the tumor response to treatment and limit normal tissue toxicity. A major limitation in that regard is the growing occurrence of intrinsic or acquired treatment resistance in advanced cases. In this review, we will discuss how overexpression of efflux pumps, perturbation of apoptosis-related factors, increased expression of antioxidants, glucose metabolism, metallotheionein expression, increased DNA repair, cancer stem cells, epithelial-mesenchymal transition, non-coding RNA and the tumour microenvironment contribute towards resistance of HNC to chemotherapy and/or radiotherapy. These mechanisms have been investigated for years and been exploited for therapeutic gain in resistant patients, paving the way to the development of new promising drugs. Since in vitro studies on resistance requires a suitable model, we will also summarize published techniques and treatment schedules that have been shown to generate acquired resistance to chemo- and/or radiotherapy that most closely mimics the clinical scenario.
Collapse
Affiliation(s)
- Tycho de Bakker
- Radiotherapy Department, Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium (S.P.)
| | - Anouk Maes
- Radiotherapy Department, Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium (S.P.)
| | - Tatiana Dragan
- Radiotherapy Department, Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium (S.P.)
| | - Philippe Martinive
- Radiotherapy Department, Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium (S.P.)
| | - Sébastien Penninckx
- Radiotherapy Department, Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium (S.P.)
- Medical Physics Department, Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Dirk Van Gestel
- Radiotherapy Department, Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium (S.P.)
| |
Collapse
|
61
|
Geng X, Chang X, Wang X, Li S, Han G, Song Z, Hao F, Li J. Consolidation immunotherapy following concurrent chemoradiotherapy in a patient with sinonasal NUT carcinoma: a case report. Front Oncol 2024; 14:1368187. [PMID: 39711966 PMCID: PMC11659667 DOI: 10.3389/fonc.2024.1368187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 10/21/2024] [Indexed: 12/24/2024] Open
Abstract
Background Nuclear protein in testis (NUT) cancers, also known as midline cancers, tends to occur in organs near the midline, such as the nasal sinuses and mediastinum. NUT carcinoma is very rare and has a poor prognosis. Case description We report the case of a 44-year-old female patient with sinonasal NUT carcinoma who presented with a soft tissue mass in the left frontal sinus, ethmoid sinus, and left nasal cavity on computed tomography; the tumor was poorly demarcated from the left rectus medialis. After discussion with a multidisciplinary team with expertise on head and neck tumors, the patient was considered inoperable, and definitive concurrent chemoradiotherapy (CCRT) was recommended. The patient underwent CCRT followed by three cycles of consolidation chemotherapy with albumin-bound paclitaxel and nedaplatin. Subsequently, the patient underwent 16 cycles of consolidation therapy with the programmed death-1 (PD-1) inhibitor tislelizumab. The immune-related adverse events included grade 2 hypothyroidism. After CCRT, consolidation chemotherapy, and consolidation immunotherapy, the patient achieved a favorable outcome. The patient survived for 31 months, and there were no signs of recurrence or metastasis during follow-up. Conclusion At present, there is no clear consensus on the consolidation treatment plan after CCRT for sinonasal NUT cancer. We used consolidation immunotherapy for the first time and achieved good efficacy, providing an innovative and promising treatment plan for refractory sinonasal NUT cancer.
Collapse
Affiliation(s)
- Xiaotao Geng
- Department of Radiation Oncology, Weifang People’s Hospital, Weifang, China
| | - Xiaolong Chang
- Department of Radiation Oncology, Weifang People’s Hospital, Weifang, China
| | - Xiaoli Wang
- Department of Radiation Oncology, Weifang People’s Hospital, Weifang, China
| | - Shunjia Li
- Department of Radiation Oncology, Weifang People’s Hospital, Weifang, China
| | - Guiyan Han
- Department of Pathology, Weifang People’s Hospital, Weifang, China
| | - Zhiyu Song
- Department of Otolaryngology, Weifang People’s Hospital, Weifang, China
| | - Furong Hao
- Department of Radiation Oncology, Weifang People’s Hospital, Weifang, China
| | - Jianwen Li
- Department of Radiation Oncology, Weifang People’s Hospital, Weifang, China
| |
Collapse
|
62
|
Chan SWS, Al Maqrashi ZAA, Young J, Kim DH, Pond G, Meyers BM, Kartolo A. Concurrent immunotherapy and radiation in cisplatin-ineligible patients with HNSCC: a systematic review & meta-analysis. Immunotherapy 2024; 16:1227-1233. [PMID: 39641447 PMCID: PMC11758945 DOI: 10.1080/1750743x.2024.2436346] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Head and squamous cell carcinoma (HNSCC) in the locally advanced setting is challenging to treat and remains an area of significant morbidity and mortality. For patients who are cisplatin-ineligible and considered unresectable, there is no clear standard of care including the choice of radiosensitizer. METHODS OVID Medline, EMBASE, and the Cochrane Central Register of Controlled Trials were systematically searched. Randomized clinical trials (RCTs) of cisplatin-ineligible unresectable HNSCC patients who were randomized to an immune checkpoint inhibitor concurrent with radiation compared to a control arm were included. Data was pooled and assessed using a random-effects model for the meta-analysis. RESULTS Three eligible RCTs were identified and analyzed which comprised 594 patients whose median age ranged from 65-72 years. There was no difference in overall survival (pooled HR 1.02, 95% CI 0.76-1.37, p = 0.88) or PFS (pooled HR 0.92, 95% CI 0.68-1.23, p = 0.56). Grade greater than or equal to 3 adverse events were favored to be less in the immunotherapy arm. CONCLUSIONS AND RELEVANCE Immune-checkpoint inhibitors are not superior to cetuximab when used with definitive radiation in HNSCC. Further study is warranted, given its potential signal for non-inferior survival with less toxicity profile trends.
Collapse
Affiliation(s)
| | | | - Jack Young
- Department of Oncology, McMaster University, Hamilton, ON, Canada
| | - Do-Hoon Kim
- Department of Oncology, McMaster University, Hamilton, ON, Canada
| | - Gregory Pond
- Department of Oncology, McMaster University, Hamilton, ON, Canada
- Escarpment Cancer Research Institute, Juravinski Cancer Centre, Hamilton, ON, Canada
| | - Brandon M Meyers
- Department of Oncology, McMaster University, Hamilton, ON, Canada
- Escarpment Cancer Research Institute, Juravinski Cancer Centre, Hamilton, ON, Canada
| | - Adi Kartolo
- Department of Oncology, McMaster University, Hamilton, ON, Canada
- Escarpment Cancer Research Institute, Juravinski Cancer Centre, Hamilton, ON, Canada
| |
Collapse
|
63
|
Mell LK, Torres-Saavedra PA, Wong SJ, Kish JA, Chang SS, Jordan RC, Liu T, Truong MT, Winquist EW, Takiar V, Wise-Draper T, Robbins JR, Rodriguez CP, Awan MJ, Beadle BM, Henson C, Narayan S, Spencer SA, Powell S, Dunlap N, Sacco AG, Hu KS, Park HS, Bauman JE, Harris J, Yom SS, Le QT. Radiotherapy with cetuximab or durvalumab for locoregionally advanced head and neck cancer in patients with a contraindication to cisplatin (NRG-HN004): an open-label, multicentre, parallel-group, randomised, phase 2/3 trial. Lancet Oncol 2024; 25:1576-1588. [PMID: 39551064 PMCID: PMC11726348 DOI: 10.1016/s1470-2045(24)00507-2] [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: 04/09/2024] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Management of patients with locoregionally advanced head and neck squamous cell carcinoma (HNSCC) when cisplatin is contraindicated is controversial. We aimed to assess whether radiotherapy with concurrent and adjuvant durvalumab would improve outcomes compared with radiotherapy with cetuximab. METHODS NRG-HN004 was designed as an open-label, multicentre, parallel-group, randomised, phase 2/3 trial with safety lead-in conducted at 89 academic and community medical centres in North America. Eligible patients were aged 18 years or older with American Joint Committee on Cancer 8th edition stage III-IVB p16-negative HNSCC or unfavourable stage I-III p16-positive oropharyngeal or unknown primary carcinoma, who had a contraindication to cisplatin (Eastern Cooperative Oncology Group [ECOG] performance status 2, renal or hearing impairment, peripheral neuropathy, aged at least 70 years with moderate or severe comorbidity, or aged younger than 70 years with severe comorbidity). Patients were randomly assigned (2:1) by permuted block randomisation (multiples of 6) to intravenous durvalumab 1500 mg starting 2 weeks before radiotherapy then every 4 weeks starting week 2 of radiotherapy (seven cycles) or intravenous cetuximab 400 mg/m2 1 week before radiotherapy then 250 mg/m2 weekly beginning week 1 of radiotherapy (eight cycles), with intensity-modulated radiotherapy (70 Gy in 35 fractions over 7 weeks). Stratification factors were tumour and nodal stage, ECOG performance status and comorbidity, and primary site and p16 status. The phase 2 primary endpoint was progression-free survival in the intention-to-treat population. There was one prespecified interim futility analysis at 50% of progression-free survival information. If the observed hazard ratio was 1·0 or more, favouring cetuximab, early stopping would be considered. Extended follow-up analysis was post hoc. This trial is registered with ClinicalTrials.gov, NCT03258554, and is closed to enrolment. FINDINGS Following a ten-patient safety lead-in, the phase 2 trial enrolled 190 patients from March 12, 2019, to July 30, 2021, 186 of whom were randomly assigned (123 to durvalumab and 63 to cetuximab). Median age was 72 years (IQR 64-77), 30 (16%) patients were women and 156 (84%) were men. Phase 2 accrual was suspended in July 30, 2021, following an interim futility analysis, and permanently closed in Sept 1, 2022. The phase 3 part of the trial was not conducted. At a median follow-up of 2·3 years (IQR 1·9-3·1) for the extended follow-up (data cutoff July 31, 2023; post-hoc analysis), 2-year progression-free survival was 50·6% (95% CI 41·5-59·8) in the durvalumab group versus 63·7% (51·3-76·1) in the cetuximab group (hazard ratio 1·33 [95% CI 0·84-2·12]; p=0·89). Adverse events were similar in both groups. The most common grade 3-4 adverse events were dysphagia (26 [22%] of 119 patients in the durvalumab group vs 18 [30%] of 61 patients in the cetuximab group), lymphopenia (33 [28%] vs 20 [33%]), and oral mucositis (13 [11%] vs 11 [18%]). Four (3%) patients in the durvalumab group and one (2%) in the cetuximab group died from treatment-related adverse events (death not otherwise specified, laryngeal oedema, lung infection, and respiratory failure in the durvalumab group and sudden death not otherwise specified in the cetuximab group). INTERPRETATION Our findings suggest that durvalumab did not improve outcomes compared with cetuximab in patients with HNSCC with contraindications to cisplatin. Further trials are needed to define the standard of care for this population. FUNDING US National Cancer Institute and AstraZeneca.
Collapse
Affiliation(s)
- Loren K Mell
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, USA.
| | - Pedro A Torres-Saavedra
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, NCI, Bethesda, MD, USA
| | - Stuart J Wong
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Julie A Kish
- Division of Hematology Oncology, Department of Medicine, Moffitt Cancer Center, Tampa, FL, USA
| | - Steven S Chang
- Department of Otorhinolaryngology, Henry Ford Health System, Detroit, MI, USA
| | - Richard C Jordan
- NRG Oncology Biospecimen Bank, University of California San Francisco, San Francisco, CA, USA
| | - Tian Liu
- Department of Radiation Oncology, Emory University, Atlanta, GA, USA
| | - Minh Tam Truong
- Department of Radiation Oncology, Boston Medical Center, Boston, MA, USA
| | - Eric W Winquist
- Department of Oncology, London Regional Cancer Program, London, ON, Canada
| | - Vinita Takiar
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Trisha Wise-Draper
- Division of Hematology Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Jared R Robbins
- Department of Radiation Oncology, University of Arizona College of Medicine, Tucson, AZ, USA
| | | | - Musaddiq J Awan
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Beth M Beadle
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Christina Henson
- Department of Radiation Oncology, University of Oklahoma, Oklahoma City, OK, USA
| | - Samir Narayan
- Department of Radiation Oncology, Trinity Health Ann Arbor, Ann Arbor, MI, USA
| | - Sharon A Spencer
- Department of Radiation Oncology, University of Alabama Birmingham, Birmingham, AL, USA
| | | | - Neal Dunlap
- Department of Radiation Oncology, University of Louisville, Louisville, KY, USA
| | - Assuntina G Sacco
- Division of Hematology Oncology, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Kenneth Shung Hu
- Department of Radiation Oncology, New York University Langone Medical Center, New York, NY, USA
| | - Henry S Park
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Julie E Bauman
- Division of Hematology/Oncology, Department of Medicine, George Washington University and George Washington Cancer Center, Washington, DC, USA
| | - Jonathan Harris
- NRG Oncology Statistics and Data Management Center, American College of Radiology, Philadelphia, PA, USA
| | - Sue S Yom
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Quynh-Thu Le
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
64
|
Stritzke F, Held T. [Pembrolizumab in combination with radiochemotherapy in patients with locally advanced squamous cell carcinoma of the head and neck: results of the KEYNOTE-412 study and the challenges of radioimmunotherapy]. Strahlenther Onkol 2024; 200:1097-1099. [PMID: 39412597 DOI: 10.1007/s00066-024-02315-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2024] [Indexed: 11/26/2024]
Affiliation(s)
- Florian Stritzke
- Klinik für Radioonkologie und Strahlentherapie, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Deutschland.
- Nationales Centrum für Tumorerkrankungen (NCT), Im Neuenheimer Feld 460, 69120, Heidelberg, Deutschland.
- Arbeitsgruppe junge DEGRO, Deutsche Gesellschaft für Radioonkologie e. V. (DEGRO), Berlin, Deutschland.
| | - Thomas Held
- Klinik für Radioonkologie und Strahlentherapie, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Deutschland
- Nationales Centrum für Tumorerkrankungen (NCT), Im Neuenheimer Feld 460, 69120, Heidelberg, Deutschland
| |
Collapse
|
65
|
Kiyota N. Patients with head and neck cancer unfit for cisplatin-what next? Lancet Oncol 2024; 25:1513-1514. [PMID: 39551067 DOI: 10.1016/s1470-2045(24)00597-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Affiliation(s)
- Naomi Kiyota
- Kobe University Hospital, Cancer Center, Kobe, Hyogo, Japan.
| |
Collapse
|
66
|
Kwong DLW. Locoregionally advanced nasopharyngeal carcinoma: integrating immunotherapy into definitive treatment. Lancet Oncol 2024; 25:1511-1513. [PMID: 39522540 DOI: 10.1016/s1470-2045(24)00570-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
|
67
|
Ellsworth SG, Ross A, Shiue KRY, Murthy P, Byrne-Steel ML, Patel R, Zellars RC, Kong FMS, Miller A, Russ KA, Lotze MT. Survey of Changes in Absolute Lymphocyte Counts and Peripheral Immune Repertoire Diversity after External Beam Radiotherapy. Radiat Res 2024; 202:837-846. [PMID: 39472998 DOI: 10.1667/rade-24-00010.1] [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: 01/05/2024] [Accepted: 10/16/2024] [Indexed: 12/07/2024]
Abstract
Radiation-induced lymphopenia (RIL) is associated with worse outcomes in patients with multiple solid tumors. Hypofractionated radiation therapy (HFRT) reduces RIL compared with conventionally fractionated radiation therapy (CFRT). However, fractionation effects on immune repertoire (IR) diversity are unknown. RNA-based T- and B-cell receptor sequencing was performed on peripheral lymphocytes collected prospectively before radiation therapy and <4 weeks after the final radiation fraction. Patients received CFRT (≤3 Gy/day × ≥10 days ± chemotherapy, n = 13) or HFRT (≥5 Gy/day × ≤5 days, n = 10), per institutional standards of care. Immune repertoire diversity parameters analyzed were number of unique CDR3 receptors (uCDR3), Shannon entropy, and sample clonality (percentage of all receptors represented by the top 10 clones). RIL was severe with concurrent chemotherapy (median %Δ ALC -58.8%, -12.5%, and -28.6% in patients treated with CFRT and chemo, CFRT alone, and HFRT, respectively). CFRT and concurrent chemotherapy was associated with more severe diversity restriction in all examined parameters than either HFRT or CFRT alone. Increased immune repertoire diversity despite decreased ALC was more common in patients treated with HFRT than CFRT and significantly less common in patients treated with concurrent chemotherapy (P < 0.001). Radiation-induced changes in immune repertoire diversity are variably reflected in the peripheral ALC. Both HFRT and CFRT caused RIL, but HFRT was associated with improved immune repertoire diversity despite RIL. The addition of chemotherapy may potentiate radiation-induced restriction in immune repertoire diversity. As immune repertoire diversity is associated with response to immunotherapy, these findings may have implications for radiation therapy/chemotherapy/immunotherapy combinations. Further studies are required to understand the relationship between radiation, circulating lymphocyte populations, immune repertoire diversity and response to treatment.
Collapse
Affiliation(s)
- Susannah G Ellsworth
- Department of Radiation Oncology, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Alison Ross
- Department of Radiation Oncology, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Kevin R Y Shiue
- Department of Radiation Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Pranav Murthy
- Department of Radiation Oncology, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania
| | | | - Ravi Patel
- Department of Radiation Oncology, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Richard C Zellars
- Department of Radiation Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Feng-Ming Spring Kong
- Department of Clinical Oncology, Hong Kong University School of Clinical Medicine, Pokfulam Hong Kong SAR, China
| | - Amy Miller
- Department of Radiation Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Kristen A Russ
- Department of Radiation Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Michael T Lotze
- Department of Radiation Oncology, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania
- Departments of Surgery, Immunology and Bioengineering; University of Pittsburgh; Pittsburgh, Pennsylvania
| |
Collapse
|
68
|
Morgenthaler J, Trommer M, Khor R, Wada M, Bahig H, Garden AS, Thai A, Gan H, Fokas E, Ping Ng S. Can we safely de-escalate HPV + oropharyngeal cancers? - A review of current practices and novel approaches. Oral Oncol 2024; 159:107089. [PMID: 39509801 DOI: 10.1016/j.oraloncology.2024.107089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
Oropharyngeal carcinomas linked to high-risk types of human papillomavirus (HPV+OPC) as a distinct tumor entity, have a better prognosis than HPV-OPC. Current treatment approaches do not differentiate between HPV-positive and negative disease, but ongoing studies are exploring de-escalation strategies, aiming to reduce therapy-related morbidity and improve patient quality of life, particularly focusing on reducing late effects from radiotherapy.We performed a literature search for both published and ongoing clinical trials and critically discussed the presented concepts and results. Those include reduction in radiotherapy dose or volume, omission or modification of concomitant chemotherapy/immunotherapy, usage of induction chemotherapy and utilization of advanced molecular and imaging biomarkers and radiomics for selected subgroups of HPV+OPC patients. While promising data have been reported from various Phase II trials, evidence from Phase III de-escalation trials has failed to demonstrate improved outcomes. Therefore, further data and an improved risk stratification are required before de-escalated radiation treatments can be recommended outside of clinical trials.The review aims to outline current de-escalation strategies and future possibilities for enhancing patient outcomes in HPV+OPC.
Collapse
Affiliation(s)
- Janis Morgenthaler
- Department of Radiation Oncology, Olivia Newton-John Cancer Wellness and Research Centre, Austin Health, Melbourne, Australia; Department of Radiation Oncology, Cyberknife and Radiotherapy, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| | - Maike Trommer
- Department of Radiation Oncology, Olivia Newton-John Cancer Wellness and Research Centre, Austin Health, Melbourne, Australia; Department of Radiation Oncology, Cyberknife and Radiotherapy, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Richard Khor
- Department of Radiation Oncology, Olivia Newton-John Cancer Wellness and Research Centre, Austin Health, Melbourne, Australia
| | - Morikatsu Wada
- Department of Radiation Oncology, Olivia Newton-John Cancer Wellness and Research Centre, Austin Health, Melbourne, Australia
| | - Houda Bahig
- Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Adam S Garden
- Department of Radiation Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Alesha Thai
- Department of Medical Oncology, Olivia Newton-John Cancer Wellness and Research Centre, Austin Health, Melbourne, Victoria, Australia
| | - Hui Gan
- Department of Medical Oncology, Olivia Newton-John Cancer Wellness and Research Centre, Austin Health, Melbourne, Victoria, Australia
| | - Emmanouil Fokas
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Sweet Ping Ng
- Department of Radiation Oncology, Olivia Newton-John Cancer Wellness and Research Centre, Austin Health, Melbourne, Australia
| |
Collapse
|
69
|
von Witzleben A, Doescher J, Hoffmann TK, Laban S. [Immunotherapy for head and neck tumors : Updates from the 2024 ASCO Annual Meeting]. HNO 2024; 72:850-856. [PMID: 39466338 DOI: 10.1007/s00106-024-01524-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] [Accepted: 09/24/2024] [Indexed: 10/30/2024]
Abstract
Immunotherapeutic approaches are now established in the treatment of various tumor entities, including head and neck squamous cell carcinoma (HNSCC). PD‑1 antibodies are currently approved for HNSCC with palliative intent but are increasingly being investigated in studies with curative objectives, e.g., as neoadjuvant therapy. At ASCO 2024, particular focus was placed on combinations of immunotherapy with therapeutic vaccines for human papillomavirus (HPV)-induced tumors. Moreover, the question of which patients benefit most from immunotherapy remains unresolved. The growing significance of PD-L1 expression, measured by the combined positive score (CPS), is becoming increasingly evident. This article summarizes the latest relevant findings from the largest international cancer congress, the American Society of Clinical Oncology (ASCO) 2024 Annual Meeting.
Collapse
Affiliation(s)
- Adrian von Witzleben
- Kopf-Hals-Tumorzentrum des Comprehensive Cancer Center Ulm, Klinik für Hals-Nasen-Ohrenheilkunde und Kopf-Hals-Chirurgie, Universitätsklinik Ulm, Frauensteige 12, 89070, Ulm, Deutschland.
| | - Johannes Doescher
- Klinik für Hals-Nasen-Ohrenheilkunde, Universitätsklinikum Augsburg, Augsburg, Deutschland
| | - Thomas K Hoffmann
- Kopf-Hals-Tumorzentrum des Comprehensive Cancer Center Ulm, Klinik für Hals-Nasen-Ohrenheilkunde und Kopf-Hals-Chirurgie, Universitätsklinik Ulm, Frauensteige 12, 89070, Ulm, Deutschland
| | - Simon Laban
- Kopf-Hals-Tumorzentrum des Comprehensive Cancer Center Ulm, Klinik für Hals-Nasen-Ohrenheilkunde und Kopf-Hals-Chirurgie, Universitätsklinik Ulm, Frauensteige 12, 89070, Ulm, Deutschland
| |
Collapse
|
70
|
Liu SL, Li XY, Yang JH, Wen DX, Guo SS, Liu LT, Li YF, Luo MJ, Xie SY, Liang YJ, Sun XS, Yang ZC, Lv XF, Luo DH, Li JB, Liu Q, Wang P, Guo L, Mo HY, Sun R, Yang Q, Lan KQ, Jia GD, Li R, Zhao C, Xu RH, Chen QY, Tang LQ, Mai HQ. Neoadjuvant and adjuvant toripalimab for locoregionally advanced nasopharyngeal carcinoma: a randomised, single-centre, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol 2024; 25:1563-1575. [PMID: 39522541 DOI: 10.1016/s1470-2045(24)00504-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Patients with locoregionally advanced nasopharyngeal carcinoma with a high pretreatment plasma concentration of Epstein-Barr virus (EBV) DNA remain at high risk for recurrence after concurrent chemoradiotherapy. This study aimed to compare the efficacy and safety of neoadjuvant-adjuvant treatment with the PD-1 inhibitor toripalimab and concurrent chemoradiotherapy versus placebo and concurrent chemoradiotherapy in patients with locoregionally advanced nasopharyngeal carcinoma. METHODS This randomised, single-centre, double-blind, placebo-controlled, phase 2 trial was conducted at Sun Yat-sen University Cancer Centre in Guangzhou, China. Adult patients (aged 18-65 years) with newly diagnosed high-risk stage III-IVa locoregionally advanced nasopharyngeal carcinoma, with a pretreatment plasma EBV DNA concentration of at least 1500 copies per mL and an Eastern Cooperative Oncology Group performance score of 0-1, were eligible. Patients were randomly assigned (2:1) using an interactive web response system (block size of six), stratified by TNM stage (III vs IVa), to neoadjuvant toripalimab (240 mg intravenously) or placebo once every 2 weeks for two cycles, followed by concurrent cisplatin (100 mg/m2 intravenously) on days 1, 22, and 43 during intensity-modulated radiotherapy and adjuvant toripalimab (240 mg intravenously) or placebo once every 3 weeks for up to eight cycles. The primary endpoint was 2-year progression-free survival in the intention-to-treat population. This study was registered with ClinicalTrials.gov, NCT03925090, and is closed to enrolment; follow-up is ongoing. FINDINGS Between Dec 6, 2019, and Dec 9, 2021, 150 patients were enrolled and randomly assigned to the toripalimab group (n=100) or placebo group (n=50). 115 (77%) patients were male and 35 (23%) were female. As of data cutoff (May 31, 2024), median follow-up for progression-free survival was 37·8 months (IQR 34·2-46·5) for the intention-to-treat population analyses. 2-year progression-free survival was higher in the toripalimab group (92·0% [95% CI 86·7-97·3]) than in the placebo group (74·0% [61·8-86·2]; stratified hazard ratio 0·40 [95% CI 0·18-0·89]; log-rank p=0·019). The most common grade 3 or worse acute adverse events (occurring within 1 year of randomisation) were leukopenia (40 [40%] of 99 patients in the toripalimab group vs 22 [44%] of 50 patients in the placebo group), mucositis (28 [28%] vs ten [20%]), neutropenia (17 [17%] vs nine [18%]), anaemia (16 [16%] vs five [10%]), and weight loss (12 [12%] vs six [12%]). The most common grade 3 or worse late adverse events (occurring >1 year after randomisation) was auditory or hearing loss (eight [8%] vs four [8%]). Immune-mediated adverse events of grade 3 or worse occurred in ten (10%) patients only in the toripalimab group. One (2%) of 50 patients in the placebo group died due to septic shock caused by bacteraemia considered not treatment related. There were no treatment-related deaths in the toripalimab group. INTERPRETATION Our findings suggested that a so-called sandwich approach involving toripalimab (in the neoadjuvant and adjuvant phases) combined with concurrent chemoradiotherapy could be a highly promising therapy for the treatment of locoregionally advanced nasopharyngeal carcinoma. Phase 3 non-inferiority trials are warranted comparing neoadjuvant and adjuvant toripalimab versus cisplatin plus gemcitabine neoadjuvant chemotherapy combined with concurrent chemoradiotherapy. FUNDING National Key Research and Development Program of China, National Natural Science Foundation of China, Guangdong Basic and Applied Basic Research Foundation, Science and Technology Program of Guangzhou, Sun Yat-sen University Clinical Research 5010 Program, Innovative Research Team of High-level Local Universities in Shanghai, Postdoctoral Innovative Talent Support Program, Planned Science and Technology Project of Guangdong Province, Key Youth Teacher Cultivating Program of Sun Yat-sen University, and Fundamental Research Funds for the Central Universities. TRANSLATION For the Chinese translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Sai-Lan Liu
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiao-Yun Li
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jin-Hao Yang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Dong-Xiang Wen
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shan-Shan Guo
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li-Ting Liu
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yi-Fu Li
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Mei-Juan Luo
- Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Si-Yi Xie
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yu-Jing Liang
- Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xue-Song Sun
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhen-Chong Yang
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiao-Fei Lv
- Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Dong-Hua Luo
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ji-Bin Li
- Clinical Trials Center, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qing Liu
- Clinical Trials Center, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Pan Wang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ling Guo
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hao-Yuan Mo
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rui Sun
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qi Yang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Kai-Qi Lan
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Guo-Dong Jia
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ru Li
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chong Zhao
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qiu-Yan Chen
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lin-Quan Tang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hai-Qiang Mai
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
71
|
Orland MD, Ullah F, Yilmaz E, Geiger JL. Immunotherapy for Head and Neck Squamous Cell Carcinoma: Present and Future Approaches and Challenges. JCO Oncol Pract 2024; 20:1588-1595. [PMID: 38709998 DOI: 10.1200/op.24.00041] [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: 01/16/2024] [Revised: 03/22/2024] [Accepted: 04/02/2024] [Indexed: 05/08/2024] Open
Abstract
Despite significant progress and improving outcomes in the management of head and neck squamous cell carcinoma (HNSCC), there are few effective treatment options for patients with recurrent or metastatic head and neck squamous cell carcinoma. The advent of immune checkpoint inhibitors has changed the treatment algorithm of head and neck squamous cell carcinoma and are approved in the frontline setting for recurrent and metastatic (R/M) head and neck squamous cell carcinomas. Although promising for some patients, most patients with R/M HNSCC do not derive clinical benefit from currently approved checkpoint inhibitors. Many studies are underway to identify the patient population that would benefit the most from immunotherapy as well as postimmunotherapy treatment failures, including novel combinations of immunomodulatory therapies. In this review, we summarize the clinical development of all major clinical trials of immunotherapy in HNSCC.
Collapse
Affiliation(s)
- Mark D Orland
- Department of Internal Medicine, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Fauzia Ullah
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Emrullah Yilmaz
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Jessica L Geiger
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
72
|
Li HX, Gong YW, Yan PJ, Xu Y, Qin G, Wen WP, Teng FY. Revolutionizing head and neck squamous cell carcinoma treatment with nanomedicine in the era of immunotherapy. Front Immunol 2024; 15:1453753. [PMID: 39676875 PMCID: PMC11638222 DOI: 10.3389/fimmu.2024.1453753] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 11/05/2024] [Indexed: 12/17/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a prevalent malignant tumor globally. Despite advancements in treatment methods, the overall survival rate remains low due to limitations such as poor targeting and low bioavailability, which result in the limited efficacy of traditional drug therapies. Nanomedicine is considered to be a promising strategy in tumor therapy, offering the potential for maximal anti-tumor effects. Nanocarriers can overcome biological barriers, enhance drug delivery efficiency to targeted sites, and minimize damage to normal tissues. Currently, various nano-carriers for drug delivery have been developed to construct new nanomedicine. This review aims to provide an overview of the current status of HNSCC treatment and the necessity of nanomedicine in improving treatment outcomes. Moreover, it delves into the research progress of nanomedicine in HNSCC treatment, with a focus on enhancing radiation sensitivity, improving the efficacy of tumor immunotherapy, effectively delivering chemotherapy drugs, and utilizing small molecule inhibitors. Finally, this article discussed the challenges and prospects of applying nanomedicine in cancer treatment.
Collapse
Affiliation(s)
- Hong-Xia Li
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Otolaryngology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Department of Endocrinology and Metabolism, Luzhou, Sichuan, China
| | - Yu-Wen Gong
- Department of Otolaryngology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Pi-Jun Yan
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Department of Endocrinology and Metabolism, Luzhou, Sichuan, China
| | - Yong Xu
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Department of Endocrinology and Metabolism, Luzhou, Sichuan, China
| | - Gang Qin
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wei-Ping Wen
- Department of Otolaryngology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Fang-Yuan Teng
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Department of Endocrinology and Metabolism, Luzhou, Sichuan, China
| |
Collapse
|
73
|
Mowery YM, Ballman KV, Hong AM, Schuetze SM, Wagner AJ, Monga V, Heise RS, Attia S, Choy E, Burgess MA, Bae S, Pryor DI, Van Tine BA, Tinoco G, Chmielowski B, Freeman C, Gronchi A, Meyer CF, Dickson MA, Hartner L, Davis LE, Powers BC, Moding EJ, Weinhold KJ, van de Rijn M, Brigman BE, Riedel RF, Kirsch DG. Safety and efficacy of pembrolizumab, radiation therapy, and surgery versus radiation therapy and surgery for stage III soft tissue sarcoma of the extremity (SU2C-SARC032): an open-label, randomised clinical trial. Lancet 2024; 404:2053-2064. [PMID: 39547252 PMCID: PMC11842127 DOI: 10.1016/s0140-6736(24)01812-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/05/2024] [Accepted: 08/28/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Approximately half of patients with localised, high-risk soft tissue sarcoma of the extremity develop metastases. We aimed to assess whether the addition of pembrolizumab to preoperative radiotherapy and surgery would improve disease-free survival. METHODS We completed an open-label, randomised clinical trial in patients with grade 2 or 3, stage III undifferentiated pleomorphic sarcoma or dedifferentiated or pleomorphic liposarcoma of the extremity and limb girdle. Patients were enrolled at 20 academic institutions in Australia, Canada, Italy, and the USA. Patients were randomly assigned to preoperative radiotherapy then surgery (control group) or preoperative pembrolizumab with radiotherapy (initiated 1-14 days after the first dose of pembrolizumab) then surgery and postoperative pembrolizumab (experimental group). Pembrolizumab (200 mg intravenously every 3 weeks) was administered as three neoadjuvant cycles (before, during, and after radiotherapy) and 14 or less adjuvant cycles. Primary endpoint was disease-free survival. This study is registered with ClincialTrials.gov (NCT03092323). FINDINGS Between Nov 18, 2017, and Nov 14, 2023, 143 participants were randomly assigned to treatment. A modified intention-to-treat analysis of 127 patients with median follow-up of 43 months showed that the experimental group (n=64) had significantly longer disease-free survival than the control group (n=63; log-rank one-sided p=0·035; hazard ratio [HR] 0·61; 90% CI 0·39-0·96). The 2-year disease-free survival increased by 15% with addition of pembrolizumab: 52% (90% CI 42-64) and 67% (90% CI 58-78) for the control and experimental groups, respectively. Disease-free survival was similarly improved with pembrolizumab for the intention-to-treat patient population (HR 0·61 [90% CI 0·39-0·95]). Grade 3 or higher adverse events occurred more frequently in the experimental group (56%) than the control group (31%). INTERPRETATION Addition of pembrolizumab to preoperative radiotherapy and surgery improves disease-free survival for patients with stage III undifferentiated pleomorphic sarcoma and pleomorphic or dedifferentiated liposarcoma of the extremity, which establishes a promising new treatment option for these patients. FUNDING Stand Up to Cancer and Merck Sharp & Dohme.
Collapse
Affiliation(s)
- Yvonne M Mowery
- Department of Radiation Oncology, UPMC Hillman Cancer Center-University of Pittsburgh, Pittsburgh, PA, USA; Department of Radiation Oncology, Duke University, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA
| | - Karla V Ballman
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Angela M Hong
- Department of Radiation Oncology, Chris O'Brien Lifehouse-Royal Prince Alfred Hospital, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Scott M Schuetze
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Andrew J Wagner
- Sarcoma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Varun Monga
- Division of Hematology and Oncology, Department of Medicine, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA; Division of Hematology and Oncology, Department of Medicine, UCSF Helen Diller Comprehensive Cancer Center, San Francisco, CA, USA
| | - Rachel S Heise
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Steven Attia
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Edwin Choy
- Division of Hematology Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Melissa A Burgess
- Division of Hematology and Oncology, Department of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Susie Bae
- Department of Medical Oncology, Peter MacCallum Cancer Centre and Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - David I Pryor
- Department of Radiation Oncology, Princess Alexandra Hospital and Queensland University of Technology, Brisbane, QLD, Australia
| | - Brian A Van Tine
- Division of Oncology, Department of Medicine, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Gabriel Tinoco
- Division of Medical Oncology, Department of Medicine, The Ohio State University, Arthur G James Comprehensive Cancer Center, Columbus, OH, USA
| | - Bartosz Chmielowski
- Division of Hematology and Medical Oncology, Department of Medicine, Jonsson Comprehensive Cancer Center, The University of California Los Angeles, Los Angeles, CA, USA
| | - Carolyn Freeman
- Department of Radiation Oncology, McGill University Health Centre, Montreal, QC, Canada
| | - Alessandro Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Christian F Meyer
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Mark A Dickson
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lee Hartner
- Division of Hematology and Medical Oncology, Department of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Lara E Davis
- Division of Hematology and Medical Oncology, Department of Medicine, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Benjamin C Powers
- Division of Medical Oncology, Department of Internal Medicine, The University of Kansas Cancer Center, Kansas City, KS, USA
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University, Palo Alto, CA, USA
| | - Kent J Weinhold
- Department of Surgery, Duke University, Durham, NC, USA; Department of Immunology, Duke University, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA
| | - Matt van de Rijn
- Department of Pathology, Stanford University, Palo Alto, CA, USA
| | - Brian E Brigman
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA
| | - Richard F Riedel
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA
| | - David G Kirsch
- Department of Radiation Oncology, Duke University, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA; Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
74
|
Chawrylak K, Homa-Mlak I, Mazurek M, Płecha E, Brzozowska A, Małecka-Massalska T, Mlak R. MiR-22-3p as a promising predictor of nutritional deficiencies in patients with head and neck cancer subjected to intensity-modulated radiation therapy. Sci Rep 2024; 14:28120. [PMID: 39548174 PMCID: PMC11568149 DOI: 10.1038/s41598-024-79641-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024] Open
Abstract
Head and neck cancer (HNC) is the seventh most common cancer globally, with 20-60% of patients experiencing nutritional deficiencies. Recent studies indicate that microRNAs (miRNAs) may serve as molecular markers for malnutrition. This study evaluated miR-22-3p as a potential predictor of nutritional deficiencies and a prognostic factor in HNC patients undergoing intensity-modulated radiation therapy (IMRT). From 2014 to 2017, fifty-six advanced HNC patients at the Medical University of Lublin received IMRT, with miR-22-3p levels measured from peripheral blood before treatment. Statistical analysis using MedCalc 15.8 revealed that underweight patients had significantly lower miR-22-3p expression compared to non-underweight patients (0.89 vs. 2.47; p = 0.0233). Moderately or severely malnourished patients also showed reduced miR-22-3p levels compared to well-nourished individuals (1.42 vs. 11.04; p = 0.026). Additionally, patients with critical weight loss (CWL) had significantly lower miR-22-3p levels than those without CWL (0.96 vs. 4.91; p = 0.0015). Weak correlations were found between miR-22-3p levels, cancer stage, body mass index (BMI), and C-reactive protein (CRP), with lower miR-22-3p levels linked to advanced tumor stages and higher CRP levels. This study suggests miR-22-3p as a biomarker for nutritional deficiency risk in HNC patients, though further research is needed to validate its predictive capacity.
Collapse
Affiliation(s)
- Katarzyna Chawrylak
- Student Scientific Group, Department of Human Physiology, Medical University of Lublin, Lublin, 20-080, Poland.
- Department of Surgical Oncology, Medical University of Lublin, Radziwiłłowska 13 St, Lublin, 20-080, Poland.
| | - Iwona Homa-Mlak
- Department of Human Physiology of Chair of Preclinical Sciences, Medical University of Lublin, Lublin, 20-080, Poland
| | - Marcin Mazurek
- Department of Human Physiology of Chair of Preclinical Sciences, Medical University of Lublin, Lublin, 20-080, Poland
| | - Ewelina Płecha
- Student Scientific Group, Department of Human Physiology, Medical University of Lublin, Lublin, 20-080, Poland
| | - Anna Brzozowska
- II Department of Radiotherapy, Center of Oncology of the Lublin Region St. John of Dukla, Lublin, 20-090, Poland
| | - Teresa Małecka-Massalska
- Department of Human Physiology of Chair of Preclinical Sciences, Medical University of Lublin, Lublin, 20-080, Poland
| | - Radosław Mlak
- Department of Laboratory Diagnostics, Medical University of Lublin, Doktora Witolda Chodźki 1 Str, Lublin, 20-093, Poland
| |
Collapse
|
75
|
Bhandari C, Moffat A, Fakhry J, Malkoochi A, Nguyen A, Trinh B, Hoyt K, Story MD, Hasan T, Obaid G. A single photodynamic priming protocol augments delivery of ⍺-PD-L1 mAbs and induces immunogenic cell death in head and neck tumors. Photochem Photobiol 2024; 100:1647-1658. [PMID: 37818742 PMCID: PMC11006828 DOI: 10.1111/php.13865] [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: 08/14/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/13/2023]
Abstract
Photodynamic priming (PDP) leverages the photobiological effects of subtherapeutic photodynamic therapy (PDT) regimens to modulate the tumor vasculature and stroma. PDP also sensitizes tumors to secondary therapies, such as immunotherapy by inducing a cascade of molecular events, including immunogenic cell death (ICD). We and others have shown that PDP improves the delivery of antibodies, among other theranostic agents. However, it is not known whether a single PDP protocol is capable of both inducing ICD in vivo and augmenting the delivery of immune checkpoint inhibitors. In this rapid communication, we show for the first time that a single PDP protocol using liposomal benzoporphyrin derivative (Lipo-BPD, 0.25 mg/kg) with 690 nm light (75 J/cm2, 100 mW/cm2) simultaneously doubles the delivery of ⍺-PD-L1 antibodies in murine AT-84 head and neck tumors and induces ICD in vivo. ICD was observed as a 3-11 fold increase in tumor cell exposure of damage-associated molecular patterns (Calreticulin, HMGB1, and HSP70). These findings suggest that this single, highly translatable PDP protocol using clinically relevant Lipo-BPD holds potential for improving immunotherapy outcomes in head and neck cancer. It can do so by simultaneously overcoming physical barriers to the delivery of immune checkpoint inhibitors, and biochemical barriers that contribute to immunosuppression.
Collapse
Affiliation(s)
- Chanda Bhandari
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Azophi Moffat
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - John Fakhry
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Ashritha Malkoochi
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Austin Nguyen
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Brian Trinh
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Kenneth Hoyt
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
- Present Address: Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Michael D. Story
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Girgis Obaid
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
76
|
Goetz JW, Rabinowits G, Kalman N, Villa A. A Review of Immunotherapy for Head and Neck Cancer. J Dent Res 2024; 103:1185-1196. [PMID: 39370694 DOI: 10.1177/00220345241271992] [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] [Indexed: 10/08/2024] Open
Abstract
The introduction of immune checkpoint inhibitors (ICIs) to oncological care has transformed the management of various malignancies, including head and neck squamous cell carcinoma (HNSCC), offering improved outcomes. The first-line treatment of recurrent and malignant HNSCC for many years was combined platinum, 5-fluorouracil, and cetuximab. Recently, the ICI pembrolizumab was approved as a first-line treatment, with or without chemotherapy, based on tumor and immune cell percentage of programmed-death ligand 1 (PD-L1). Multiple head and neck (HN) cancer trials have subsequently explored immunotherapies in combination with surgery, chemotherapy, and/or radiation. Immunotherapy regimens may be personalized by tumor biomarker, including PD-L1 content, tumor mutational burden, and microsatellite instability. However, further clinical trials are needed to refine biomarker-driven protocols and standardize pathological methods to guide combined regimen timing, sequencing, and deescalation. Gaps remain for protocols using immunotherapy to reverse oral premalignant lesions, particularly high-risk leukoplakias. A phase II nonrandomized controlled trial, using the ICI nivolumab, showed a 2-y cancer-free survival of 73%, although larger trials are needed. Guidelines are also needed to standardize the role of dental evaluation and care before, during, and after immunotherapy, specifically in regard to oral immune-related adverse events and their impact on cancer recurrence. Standardized diagnostic and oral care coordination strategies to close these gaps are needed to ensure continued success of HN cancer immunotherapy.
Collapse
Affiliation(s)
- J W Goetz
- Oral Medicine, Oral Oncology and Dentistry, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
| | - G Rabinowits
- Department of Head and Neck - Endocrine Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - N Kalman
- Oral Medicine, Oral Oncology and Dentistry, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
| | - A Villa
- Oral Medicine, Oral Oncology and Dentistry, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
| |
Collapse
|
77
|
Abouali H, Przedborski M, Kohandel M, Poudineh M. Investigating nano-sized tumor-derived extracellular vesicles in enhancing anti-PD-1 immunotherapy. NANOSCALE 2024; 16:19062-19073. [PMID: 39319505 DOI: 10.1039/d4nr00729h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Anti-PD1 immune checkpoint blockade (ICB) has shown promising results for treating several aggressive cancers, enhancing patient survival rates. The variability in clinical response to anti-PD1 ICB is thought to be driven by patient-specific biology and heterogeneity within the tumor microenvironment. Tumor-derived extracellular vesicles (TDEVs), nano-sized particles released from tumor cells, can modulate the tumor microenvironment, leading to immunosuppression and tumor progression. Hence, TDEVs may contribute to the variability in treatment response and play a crucial role in the failure of anti-PD1 immunotherapy. In this study, we develop a systems biology approach to interrogate the role of TDEVs on the response dynamics for anti-PD1 blockade. Our results suggest that the detection and profiling of TDEVs can help screen patients for anti-PD-1 immunotherapy. Moreover, the results in this study suggest that TDEVs and IL-12 can potentially be liquid biopsy biomarkers to profile patient response to anti-PD1 ICB and tailor patient-specific treatment protocols. Importantly, the methodology is generalizable to other types of cancer immunotherapies. Therefore, the collection of patient-specific liquid biopsy data, and the implementation of those data into the systems biology framework, may offer the opportunity to discover new biomarkers for patient drug screening and enable the continuous monitoring of patient response to treatment and adaptation of patient-specific immunotherapy treatment protocols to overcome therapeutic resistance.
Collapse
Affiliation(s)
- Hesam Abouali
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Ontario, Canada.
| | - Michelle Przedborski
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada.
| | - Mohammad Kohandel
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada.
| | - Mahla Poudineh
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Ontario, Canada.
| |
Collapse
|
78
|
Lee AM, Weaver AN, Acosta P, Harris L, Bowles DW. Review of Current and Future Medical Treatments in Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2024; 16:3488. [PMID: 39456583 PMCID: PMC11506581 DOI: 10.3390/cancers16203488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/03/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a complex cancer requiring a multidisciplinary approach. For patients with locally or regionally advanced disease, surgery and/or radiation are the cornerstones of definitive treatment. Medical therapy plays an important adjunct role in this setting, typically consisting of a platinum-based regimen given as induction, concurrent, or adjuvant treatment. While relapsed/metastatic HNSCC has historically been a difficult-to-treat disease with poor outcomes, options have considerably improved with the incorporation of biologics and immune checkpoint inhibitors. Clinical trials are ongoing to investigate novel approaches, including new and combination immunotherapies, targeted therapies, therapeutic vaccines, antibody-drug conjugates, and cellular therapies. The results thus far have been mixed, highlighting the knowledge gaps that continue to challenge the medical oncologist treating HNSCC. Here, we present the most updated and broad review of the current treatment landscape in both locoregional and metastatic HNSCC and discuss the expansive future medical therapies under investigation.
Collapse
Affiliation(s)
- Aaron M. Lee
- Division of Medical Oncology, University of Colorado School of Medicine, Aurora, CO 80045, USA; (A.M.L.); (A.N.W.)
| | - Alice N. Weaver
- Division of Medical Oncology, University of Colorado School of Medicine, Aurora, CO 80045, USA; (A.M.L.); (A.N.W.)
| | - Phillip Acosta
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA;
| | - Lauren Harris
- University of Colorado School of Medicine, Aurora, CO 80045, USA;
| | - Daniel W. Bowles
- Division of Medical Oncology, University of Colorado School of Medicine, Aurora, CO 80045, USA; (A.M.L.); (A.N.W.)
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO 80045, USA
| |
Collapse
|
79
|
Lin JY, Lu ZJ, Li SC, Luo DH, Liu T, Zhang WR, Yang ZC, Mo HY, Mai HQ, Liu SL. Individualised cumulative cisplatin dose for locoregionally advanced nasopharyngeal carcinoma patients based on induction chemotherapy response and tumour volume. Ther Adv Med Oncol 2024; 16:17588359241286222. [PMID: 39483137 PMCID: PMC11526242 DOI: 10.1177/17588359241286222] [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: 02/17/2024] [Accepted: 09/05/2024] [Indexed: 11/03/2024] Open
Abstract
Background and objectives To evaluate the prognostic value of an integrated model consisting of tumour response to induction chemotherapy (IC) and gross tumour volume (GTV) after IC in nasopharyngeal carcinoma (NPC) and elucidate optimal cumulative cisplatin dose (CCD) in concurrent chemoradiotherapy (CCRT) for different subgroups. Design and methods This retrospective study enrolled 896 patients with NPC diagnosed from 2010 to 2017 receiving IC plus radiotherapy. The primary endpoint was disease-free survival (DFS). Cut-off points for GTV were combined with IC response to develop an integrated model. Propensity score matching (PSM) was used to adjust for potential confounders. Survival outcomes and acute toxicity were compared between the different CCD groups. Results Unsatisfactory IC response and large GTV after IC were correlated with poor survival outcomes; the AUC increased to 0.668 when these factors were incorporated. The integrated model classified patients into three groups. After PSM, radiotherapy alone and CCRT demonstrated similar efficacy in the low-risk group (complete response (CR)/partial response (PR) and GTV <68 cm3 after IC). In the intermediate-risk group (CR/PR but GTV ⩾68 cm3), CCD of >200 mg/m2 and 101-200 mg/m2 increased the 5-year DFS rates (83.7% vs 81.1% vs 65.3%, p = 0.042). In the high-risk group (stable disease/progressive disease and any GTV), the use of different CCDs did not result in significantly different survival outcomes (p = 0.793). Additionally, high CCD was significantly associated with increased incidence of grade 1-4 acute toxicity. Conclusion The integrated model incorporating IC response and GTV after IC demonstrates satisfactory value in risk stratification and the potential to guide individualised decision-making in CCD selection. Balancing toxicity and efficacy, RT alone seems to be the optimal treatment for patients in low-risk groups and 200 mg/m2 might be the optimal dose for intermediate-risk groups. Moreover, increasing CCD does not benefit patients in high-risk groups, and treatment options for these patients require further consideration.
Collapse
Affiliation(s)
- Jie-Yi Lin
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China
| | - Zi-Jian Lu
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China
| | - Su-Chen Li
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China
| | - Dong-Hua Luo
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China
| | - Ting Liu
- Breast Disease Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Wan-Ru Zhang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China
| | - Zhen-Chong Yang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, P.R. China
| | - Hao-Yuan Mo
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 651 Dongfeng Road East, Guangzhou 510060, P.R. China
| | - Hai-Qiang Mai
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 651 Dongfeng Road East, Guangzhou 510060, P.R. China
| | - Sai-Lan Liu
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 651 Dongfeng Road East, Guangzhou 510060, P.R. China
| |
Collapse
|
80
|
Boreel DF, Sandker GGW, Ansems M, van den Bijgaart RJE, Peters JPW, Span PN, Adema GJ, Heskamp S, Bussink J. MHC-I and PD-L1 Expression is Associated with Decreased Tumor Outgrowth and is Radiotherapy-inducible in the Murine Head and Neck Squamous Cell Carcinoma Model MOC1. Mol Imaging Biol 2024; 26:835-846. [PMID: 39009951 PMCID: PMC11436446 DOI: 10.1007/s11307-024-01934-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: 11/22/2023] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/17/2024]
Abstract
INTRODUCTION Combined radiotherapy and immune checkpoint inhibition is a potential treatment option for head and neck squamous cell carcinoma (HNSCC). Immunocompetent mouse models can help to successfully develop radio- immunotherapy combinations and to increase our understanding of the effects of radiotherapy on the tumor microenvironment for future clinical translation. Therefore, the aim of this study was to develop a homogeneous, reproducible HNSCC model originating from the Mouse Oral Cancer 1 (MOC1) HNSCC cell line, and to explore the radiotherapy-induced changes in its tumor microenvironment, using flow cytometry and PD-L1 microSPECT/CT imaging. MATERIALS AND METHODS In vivo growing tumors originating from the parental MOC1 line were used to generate single cell derived clones. These clones were screened in vitro for their ability to induce programmed cell death ligand 1 (PD-L1) and major histocompatibility complex class I (MHC-I) following IFNγ exposure. Clones with different IFNγ sensitivity were inoculated in C57BL/6 mice and assessed for tumor outgrowth. The composition of the tumor microenvironment of a stably growing (non)irradiated MOC1-derived clone was assessed by immunohistochemistry, flow cytometry and PD-L1 microSPECT/CT. RESULTS Low in vitro inducibility of MHC-I and PD-L1 by IFNγ was associated with increased tumor outgrowth of MOC1 clones in vivo. Flow cytometry analysis of cells derived from a stable in vivo growing MOC1 clone MOC1.3D5low showed expression of MHC-I and PD-L1 on several cell populations within the tumor. Upon irradiation, MHC-I and PD-L1 increased on leukocytes (CD45.2+) and cancer associated fibroblasts (CD45.2-/EpCAM-/CD90.1+). Furthermore, PD-L1 microSPECT/CT showed increased tumor uptake of radiolabeled PD-L1 antibodies with a heterogeneous spatial distribution of the radio signal, which co-localized with PD-L1+ and CD45.2+ areas. DISCUSSION PD-L1 and MHC-I inducibility by IFNγ in vitro is associated with tumor outgrowth of MOC1 clones in vivo. In tumors originating from a stably growing MOC1-derived clone, expression of these immune-related markers was induced by irradiation shown by flow cytometry on several cell populations within the tumor microenvironment such as immune cells and cancer associated fibroblasts. PD-L1 microSPECT/CT showed increased tumor uptake following radiotherapy, and autoradiography showed correlation of uptake with areas that are heavily infiltrated by immune cells. Knowledge of radiotherapy-induced effects on the tumor microenvironment in this model can help optimize timing and dosage for radio- immunotherapy combination strategies in future research.
Collapse
Affiliation(s)
- Daan F Boreel
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Geert Grooteplein Zuid 32, 6525GA, Nijmegen, The Netherlands.
- Department of Medical Imaging, Radboudumc, Geert Grooteplein 10, Nijmegen, 6525GA, The Netherlands.
| | - Gerwin G W Sandker
- Department of Medical Imaging, Radboudumc, Geert Grooteplein 10, Nijmegen, 6525GA, The Netherlands
| | - Marleen Ansems
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Geert Grooteplein Zuid 32, 6525GA, Nijmegen, The Netherlands
| | - Renske J E van den Bijgaart
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Geert Grooteplein Zuid 32, 6525GA, Nijmegen, The Netherlands
| | - Johannes P W Peters
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Geert Grooteplein Zuid 32, 6525GA, Nijmegen, The Netherlands
| | - Paul N Span
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Geert Grooteplein Zuid 32, 6525GA, Nijmegen, The Netherlands
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Geert Grooteplein Zuid 32, 6525GA, Nijmegen, The Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Radboudumc, Geert Grooteplein 10, Nijmegen, 6525GA, The Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Geert Grooteplein Zuid 32, 6525GA, Nijmegen, The Netherlands
| |
Collapse
|
81
|
Fishman D, Choe J. Immunotherapy in Head and Neck Cancer: Treatment Paradigms, Future Directions, and Questions. Surg Oncol Clin N Am 2024; 33:605-615. [PMID: 39244283 DOI: 10.1016/j.soc.2024.04.001] [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] [Indexed: 09/09/2024]
Abstract
The use of immunotherapy in head and neck squamous cell carcinoma (HNSCC)has increased treatment options for patients who may not be candidates for traditional cytotoxic chemotherapy. Recent studies have resulted in the approval of immunotherapy in the first and second line setting for recurrent/metastatic disease. Various combinations of immunotherapy with targeted therapies, monoclonal antibodies, or human papilloma virus vaccines are also being studied in recurrent/metastatic disease. Currently, programmed death-ligand 1 status is the main marker utilized to assess potential response to immunotherapy. Studies are focused on identifying additional markers, which may help better predict response to immunotherapy for HNSCC patients.
Collapse
Affiliation(s)
- Danielle Fishman
- Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue S, Nashville, TN 37232, USA; Division of Hematology/Oncology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 2220 Pierce Avenue, PRB 790, Nashville, TN 37232, USA
| | - Jennifer Choe
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 2220 Pierce Avenue, PRB 790, Nashville, TN 37232, USA.
| |
Collapse
|
82
|
Liu XY, Shang-guan HJ, Zhang W, Chen S, Luo XY. Efficacy of PD-1 inhibitor with neoadjuvant chemotherapy in hypopharyngeal and oropharyngeal cancer. Front Oncol 2024; 14:1450830. [PMID: 39381046 PMCID: PMC11458719 DOI: 10.3389/fonc.2024.1450830] [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: 06/18/2024] [Accepted: 09/02/2024] [Indexed: 10/10/2024] Open
Abstract
Objective This study is aimed to evaluate the efficacy and safety of PD-1 inhibitors combined with neoadjuvant chemotherapy in patients with locally advanced hypopharyngeal and oropharyngeal cancer prior to surgical resection. Methods This retrospective analysis included 42 patients diagnosed with locally advanced hypopharyngeal and oropharyngeal cancer. The efficacy, safety, survival, and laryngeal preservation rate were evaluated. Results A total of 42 patients were included in this retrospective analysis, of whom 28 had hypopharyngeal cancer and 14 had oropharyngeal cancer. Of the 42 patients, 14 (33.3%) achieved a pathological complete response (PCR) at the primary site, 20 (47.6%) achieved a major pathological response (MPR), and 8 (19%) had an incomplete pathological response (IPR) at the primary lesion. A PCR at both the primary site and the neck lymph nodes was observed in 9 patients (21.4%). The laryngeal preservation rate was 92.9% (26/28) in patients with hypopharyngeal cancer. The median follow-up time was 10.5 months. The median progression-free survival (PFS) was 26.42 months (95% CI, 23.416-29.424), and the median overall survival (OS) was 27.1 months (95% CI, 24.316-29.884). The 1-year PFS rate was 83.1%, and the 1-year OS rate was 85.9%. Conclusion Combination therapy with PD-1 inhibitors and neoadjuvant chemotherapy has demonstrated superior efficacy and safety as a preoperative treatment for locally advanced hypopharyngeal and oropharyngeal cancer. Notably, this treatment regimen does not increase the risk of severe postoperative complications and has shown promising results in improving laryngeal preservation rates.
Collapse
Affiliation(s)
- Xue-Ying Liu
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Han-jing Shang-guan
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wei Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang’an Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, China
| | - Shuai Chen
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Department of Otolaryngology Head and Neck Surgery, Xiamen Key Laboratory of Otolaryngology Head and Neck Surgery, Xiamen, Fujian, China
| | - Xian-Yang Luo
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Department of Otolaryngology Head and Neck Surgery, Xiamen Key Laboratory of Otolaryngology Head and Neck Surgery, Xiamen, Fujian, China
| |
Collapse
|
83
|
Belfiore MP, Nardone V, D’Onofrio I, Pirozzi M, Sandomenico F, Farese S, De Chiara M, Balbo C, Cappabianca S, Fasano M. Recurrent Versus Metastatic Head and Neck Cancer: An Evolving Landscape and the Role of Immunotherapy. Biomedicines 2024; 12:2080. [PMID: 39335592 PMCID: PMC11428618 DOI: 10.3390/biomedicines12092080] [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: 07/16/2024] [Revised: 09/01/2024] [Accepted: 09/07/2024] [Indexed: 09/30/2024] Open
Abstract
Squamous cell carcinoma of the head and neck (SCCHN) is among the ten most common cancers worldwide, with advanced SCCHN presenting with a 5-year survival of 34% in the case of nodal involvement and 8% in the case of metastatic disease. Disease-free survival at 2 years is 67% for stage II and 33% for stage III tumors, whereas 12-30% of patients undergo distant failures after curative treatment. Previous treatments often hinder the success of salvage surgery and/or reirradiation, while the standard of care for the majority of metastatic SCCHN remains palliative chemo- and immuno-therapy, with few patients eligible for locoregional treatments. The aim of this paper is to review the characteristics of recurrent SCCHN, based on different recurrence sites, and metastatic disease; we will also explore the possibilities not only of salvage surgery and reirradiation but also systemic therapy choices and locoregional treatment for metastatic SCCHN.
Collapse
Affiliation(s)
- Maria Paola Belfiore
- Diagnostic of Imaging, Department of Precision Medicine, Campania University ”L.Vanvitelli”, 80131 Naples, Italy; (V.N.); (I.D.); (M.D.C.); (S.C.)
| | - Valerio Nardone
- Diagnostic of Imaging, Department of Precision Medicine, Campania University ”L.Vanvitelli”, 80131 Naples, Italy; (V.N.); (I.D.); (M.D.C.); (S.C.)
| | - Ida D’Onofrio
- Diagnostic of Imaging, Department of Precision Medicine, Campania University ”L.Vanvitelli”, 80131 Naples, Italy; (V.N.); (I.D.); (M.D.C.); (S.C.)
| | - Mario Pirozzi
- SCDU Oncologia, “Maggiore della Carità” University Hospital, 28100 Novara, Italy;
| | - Fabio Sandomenico
- Radiology Unit, Buon Consiglio Fatebenefratelli Hospital, 80123 Naples, Italy;
| | - Stefano Farese
- Medical Oncology, Department of Precision Medicine, Campania University “L.Vanvitelli”, 80131 Naples, Italy; (S.F.); (C.B.); (M.F.)
| | - Marco De Chiara
- Diagnostic of Imaging, Department of Precision Medicine, Campania University ”L.Vanvitelli”, 80131 Naples, Italy; (V.N.); (I.D.); (M.D.C.); (S.C.)
| | - Ciro Balbo
- Medical Oncology, Department of Precision Medicine, Campania University “L.Vanvitelli”, 80131 Naples, Italy; (S.F.); (C.B.); (M.F.)
| | - Salvatore Cappabianca
- Diagnostic of Imaging, Department of Precision Medicine, Campania University ”L.Vanvitelli”, 80131 Naples, Italy; (V.N.); (I.D.); (M.D.C.); (S.C.)
| | - Morena Fasano
- Medical Oncology, Department of Precision Medicine, Campania University “L.Vanvitelli”, 80131 Naples, Italy; (S.F.); (C.B.); (M.F.)
| |
Collapse
|
84
|
Berzaghi R, Gundersen K, Dille Pedersen B, Utne A, Yang N, Hellevik T, Martinez-Zubiaurre I. Immunological signatures from irradiated cancer-associated fibroblasts. Front Immunol 2024; 15:1433237. [PMID: 39308864 PMCID: PMC11412886 DOI: 10.3389/fimmu.2024.1433237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 08/14/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction Cancer-associated fibroblasts (CAFs) are abundant and influential elements of the tumor microenvironment (TME), giving support to tumor development in multiple ways. Among other mechanisms, CAFs are important regulators of immunological processes occurring in tumors. However, CAF-mediated tumor immunomodulation in the context of radiotherapy remains poorly understood. In this study, we explore effects of radiation on CAF-derived immunoregulatory signals to the TME. Methods Primary CAF cultures were established from freshly collected human NSCLC lung tumors. CAFs were exposed to single-high or fractionated radiation regimens (1x18Gy or 3x6Gy), and the expression of different immunoregulatory cell-associated and secreted signaling molecules was analyzed 48h and 6 days after initiation of treatment. Analyses included quantitative measurements of released damage-associated molecular patterns (DAMPs), interferon (IFN) type I responses, expression of immune regulatory receptors, and secretion of soluble cytokines, chemokines, and growth factors. CAFs are able to survive ablative radiation regimens, however they enter into a stage of premature cell senescence. Results Our data show that CAFs avoid apoptosis and do not contribute by release of DAMPs or IFN-I secretion to radiation-mediated tumor immunoregulation. Furthermore, the secretion of relevant immunoregulatory cytokines and growth factors including TGF-β, IL-6, IL-10, TNFα, IL-1β, VEGF, CXCL12, and CXCL10 remain comparable between non-irradiated and radiation-induced senescent CAFs. Importantly, radiation exposure modifies the cell surface expression of some key immunoregulatory receptors, including upregulation of CD73 and CD276. Discussion Our data suggest that CAFs do not participate in the release of danger signals or IFN-I secretion following radiotherapy. The immune phenotype of CAFs and radiation-induced senescent CAFs is similar, however, the observed elevation of some cell surface immunological receptors on irradiated CAFs could contribute to the establishment of an enhanced immunosuppressive TME after radiotherapy.
Collapse
Affiliation(s)
- Rodrigo Berzaghi
- Department of Clinical Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Kristian Gundersen
- Department of Clinical Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Brede Dille Pedersen
- Department of Radiation Oncology, University Hospital of North Norway, Tromsø, Norway
| | - Amalie Utne
- Department of Radiation Oncology, University Hospital of North Norway, Tromsø, Norway
| | - Nannan Yang
- Department of Community Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Turid Hellevik
- Department of Radiation Oncology, University Hospital of North Norway, Tromsø, Norway
| | - Inigo Martinez-Zubiaurre
- Department of Clinical Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
85
|
von der Grün J, Broglie M, Guckenberger M, Balermpas P. A comprehensive and longitudinal evaluation of the different populations of lymphoid and myeloid cells in the peripheral blood of patients treated with chemoradiotherapy for head and neck cancer. Cancer Immunol Immunother 2024; 73:222. [PMID: 39235625 PMCID: PMC11377404 DOI: 10.1007/s00262-024-03810-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 08/12/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Immunotherapy provided significant survival benefits for recurrent and metastatic patients with head and neck cancer. These improvements could not be reproduced in patients treated with curative-intent chemoradiotherapy (CRT) and the optimal radio-immunotherapy (RIT) concepts have yet to be designed. Exploration and analysis of the pre-therapeutic immune status of these patients and the changes occurring during the treatment course could be crucial in rationally designing future combined treatments. METHODS Blood samples were collected from a cohort of 25 head and neck cancer patients treated with curative-intended (C)-RT prior to therapy, after the first week of treatment, and three months after treatment completion. Peripheral blood mononuclear cells (PBMCs) or all nucleated blood cells were isolated and analyzed via flow cytometry. RESULTS At baseline, patients showed reduced monocyte and lymphocyte counts compared to healthy individuals. Although overall CD8+ T-cell frequencies were reduced, the proportion of memory subsets were increased in patients. Radiotherapy (RT) treatment led to a further increase in CD8+ effector memory T-cells. Among myeloid populations, tumor-promoting subsets became less abundant after RT, in favor of pro-inflammatory cells. CONCLUSION The present study prospectively demonstrated a complex interplay and distinct longitudinal changes in the composition of lymphocytic and myeloid populations during curative (C)-RT of head and neck cancer. Further validation of this method in a larger cohort could allow for better treatment guidance and tailored incorporation of immunotherapies (IT) in the future.
Collapse
Affiliation(s)
- Jens von der Grün
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Universitäts Spital Zürich (USZ), Rämistrasse 100, 8091, Zurich, Switzerland
| | - Martina Broglie
- Department of Otorhinolaryngology-Head and Neck Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Universitäts Spital Zürich (USZ), Rämistrasse 100, 8091, Zurich, Switzerland
| | - Panagiotis Balermpas
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Universitäts Spital Zürich (USZ), Rämistrasse 100, 8091, Zurich, Switzerland.
| |
Collapse
|
86
|
Ahmad R, Barcellini A, Baumann K, Benje M, Bender T, Bragado P, Charalampopoulou A, Chowdhury R, Davis AJ, Ebner DK, Eley J, Kloeber JA, Mutter RW, Friedrich T, Gutierrez-Uzquiza A, Helm A, Ibáñez-Moragues M, Iturri L, Jansen J, Morcillo MÁ, Puerta D, Kokko AP, Sánchez-Parcerisa D, Scifoni E, Shimokawa T, Sokol O, Story MD, Thariat J, Tinganelli W, Tommasino F, Vandevoorde C, von Neubeck C. Particle Beam Radiobiology Status and Challenges: A PTCOG Radiobiology Subcommittee Report. Int J Part Ther 2024; 13:100626. [PMID: 39258166 PMCID: PMC11386331 DOI: 10.1016/j.ijpt.2024.100626] [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: 07/03/2024] [Accepted: 08/02/2024] [Indexed: 09/12/2024] Open
Abstract
Particle therapy (PT) represents a significant advancement in cancer treatment, precisely targeting tumor cells while sparing surrounding healthy tissues thanks to the unique depth-dose profiles of the charged particles. Furthermore, their linear energy transfer and relative biological effectiveness enhance their capability to treat radioresistant tumors, including hypoxic ones. Over the years, extensive research has paved the way for PT's clinical application, and current efforts aim to refine its efficacy and precision, minimizing the toxicities. In this regard, radiobiology research is evolving toward integrating biotechnology to advance drug discovery and radiation therapy optimization. This shift from basic radiobiology to understanding the molecular mechanisms of PT aims to expand the therapeutic window through innovative dose delivery regimens and combined therapy approaches. This review, written by over 30 contributors from various countries, provides a comprehensive look at key research areas and new developments in PT radiobiology, emphasizing the innovations and techniques transforming the field, ranging from the radiobiology of new irradiation modalities to multimodal radiation therapy and modeling efforts. We highlight both advancements and knowledge gaps, with the aim of improving the understanding and application of PT in oncology.
Collapse
Affiliation(s)
- Reem Ahmad
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Amelia Barcellini
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
- Clinical Department Radiation Oncology Unit, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Kilian Baumann
- Institute of Medical Physics and Radiation Protection, University of Applied Sciences Giessen, Giessen, Germany
- Marburg Ion-Beam Therapy Center, Marburg, Germany
| | - Malte Benje
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Tamara Bender
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Paloma Bragado
- Biochemistry and Molecular Biology Department, Complutense University of Madrid, Madrid, Spain
| | - Alexandra Charalampopoulou
- University School for Advanced Studies (IUSS), Pavia, Italy
- Radiobiology Unit, Development and Research Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Reema Chowdhury
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Anthony J. Davis
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Daniel K. Ebner
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - John Eley
- Department of Radiation Oncology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jake A. Kloeber
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Robert W. Mutter
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Thomas Friedrich
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | | | - Alexander Helm
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Marta Ibáñez-Moragues
- Medical Applications of Ionizing Radiation Unit, Technology Department, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Lorea Iturri
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Orsay, France
| | - Jeannette Jansen
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Miguel Ángel Morcillo
- Medical Applications of Ionizing Radiation Unit, Technology Department, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Daniel Puerta
- Departamento de Física Atómica, Molecular y Nuclear, Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Complejo Hospitalario Universitario de Granada/Universidad de Granada, Granada, Spain
| | | | | | - Emanuele Scifoni
- TIFPA-INFN - Trento Institute for Fundamental Physics and Applications, Trento, Italy
| | - Takashi Shimokawa
- National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Olga Sokol
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | | | - Juliette Thariat
- Centre François Baclesse, Université de Caen Normandie, ENSICAEN, CNRS/IN2P3, LPC Caen UMR6534, Caen, France
| | - Walter Tinganelli
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Francesco Tommasino
- TIFPA-INFN - Trento Institute for Fundamental Physics and Applications, Trento, Italy
- Department of Physics, University of Trento, Trento, Italy
| | - Charlot Vandevoorde
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Cläre von Neubeck
- Department of Particle Therapy, University Hospital Essen, University of Duisburg-Essen, Duisburg, Germany
| |
Collapse
|
87
|
Dover L, Dulaney C. PROshot: Immunotherapy for Cervical Cancer, Epidermal Growth Factor Receptor-Mutated Stage III Lung Cancer, Perioperative Chemotherapy for Esophageal Cancer, Salvage Postprostatectomy Radiation and Androgen Deprivation Therapy, and Immunotherapy for Head and Neck Cancer. Pract Radiat Oncol 2024; 14:363-367. [PMID: 39218526 DOI: 10.1016/j.prro.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 09/04/2024]
Affiliation(s)
- Laura Dover
- Department of Radiation Oncology, Ascension St. Vincent's East, Birmingham, Alabama
| | - Caleb Dulaney
- Department of Radiation Oncology, Anderson Regional Health System, Meridian, Mississippi.
| |
Collapse
|
88
|
O'Leary B, Skinner H, Schoenfeld JD, Licitra L, Le Tourneau C, Esdar C, Schroeder A, Salmio S, Psyrri A. Evasion of apoptosis and treatment resistance in squamous cell carcinoma of the head and neck. Cancer Treat Rev 2024; 129:102773. [PMID: 38878677 DOI: 10.1016/j.ctrv.2024.102773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 08/18/2024]
Abstract
Combinations of surgery, radiotherapy and chemotherapy can eradicate tumors in patients with locally advanced squamous cell carcinoma of the head and neck (LA SCCHN), but a significant proportion of tumors progress, recur, or do not respond to therapy due to treatment resistance. The prognosis for these patients is poor, thus new approaches are needed to improve outcomes. Key resistance mechanisms to chemoradiotherapy (CRT) in patients with LA SCCHN are alterations to the pathways that mediate apoptosis, a form of programmed cell death. Targeting dysregulation of apoptotic pathways represents a rational therapeutic strategy in many types of cancer, with a number of proteins, including the pro-survival B-cell lymphoma 2 family and inhibitors of apoptosis proteins (IAPs), having been identified as druggable targets. This review discusses the mechanisms by which apoptosis occurs under physiological conditions, and how this process is abnormally restrained in LA SCCHN tumor cells, with treatment strategies aimed at re-enabling apoptosis in LA SCCHN also considered. In particular, the development of, and future opportunities for, IAP inhibitors in LA SCCHN are discussed, in light of recent encouraging proof-of-concept clinical trial data.
Collapse
Affiliation(s)
| | | | | | - Lisa Licitra
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan and University of Milan, Italy
| | | | | | | | | | - Amanda Psyrri
- Attikon University Hospital, National Kapodistrian University of Athens, Greece
| |
Collapse
|
89
|
Margalit DN, Anker CJ, Aristophanous M, Awan M, Bajaj GK, Bradfield L, Califano J, Caudell JJ, Chapman CH, Garden AS, Harari PM, Helms A, Lin A, Maghami E, Mehra R, Parker L, Shnayder Y, Spencer S, Swiecicki PL, Tsai JC, Sher DJ. Radiation Therapy for HPV-Positive Oropharyngeal Squamous Cell Carcinoma: An ASTRO Clinical Practice Guideline. Pract Radiat Oncol 2024; 14:398-425. [PMID: 39078350 DOI: 10.1016/j.prro.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 07/31/2024]
Abstract
PURPOSE Human Papilloma Virus (HPV)-associated oropharyngeal squamous cell carcinoma (OPSCC) is a distinct disease from other head and neck tumors. This guideline provides evidence-based recommendations on the critical decisions in its curative treatment, including both definitive and postoperative radiation therapy (RT) management. METHODS ASTRO convened a task force to address 5 key questions on the use of RT for management of HPV-associated OPSCC. These questions included indications for definitive and postoperative RT and chemoradiation; dose-fractionation regimens and treatment volumes; preferred RT techniques and normal tissue considerations; and posttreatment management decisions. The task force did not address indications for primary surgery versus RT. Recommendations were based on a systematic literature review and created using a predefined consensus-building methodology and system for grading evidence quality and recommendation strength. RESULTS Concurrent cisplatin is recommended for patients receiving definitive RT with T3-4 disease and/or 1 node >3 cm, or multiple nodes. For similar patients who are ineligible for cisplatin, concurrent cetuximab, carboplatin/5-fluorouracil, or taxane-based systemic therapy are conditionally recommended. In the postoperative setting, RT with concurrent cisplatin (either schedule) is recommended for positive surgical margins or extranodal extension. Postoperative RT alone is recommended for pT3-4 disease, >2 nodes, or a single node >3 cm. Observation is conditionally recommended for pT1-2 disease and a single node ≤3 cm without other risk factors. For patients treated with definitive RT with concurrent systemic therapy, 7000 cGy in 33 to 35 fractions is recommended, and for patients receiving postoperative RT without positive surgical margins and extranodal extension, 5600 to 6000 cGy is recommended. For all patients receiving RT, intensity modulated RT over 3-dimensional techniques with reduction in dose to critical organs at risk (including salivary and swallowing structures) is recommended. Reassessment with positron emission tomography-computed tomography is recommended approximately 3 months after definitive RT/chemoradiation, and neck dissection is recommended for convincing evidence of residual disease; for equivocal positron emission tomography-computed tomography findings, either neck dissection or repeat imaging is recommended. CONCLUSIONS The role and practice of RT continues to evolve for HPV-associated OPSCC, and these guidelines inform best clinical practice based on the available evidence.
Collapse
Affiliation(s)
- Danielle N Margalit
- Department of Radiation Oncology, Brigham & Women's/Dana-Farber Cancer Center, Harvard Medical School, Boston, Massachusetts.
| | - Christopher J Anker
- Division of Radiation Oncology, University of Vermont Cancer Center, Burlington, Vermont
| | - Michalis Aristophanous
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Musaddiq Awan
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Gopal K Bajaj
- Department of Advanced Radiation Oncology and Proton Therapy, Inova Schar Cancer Institute, Fairfax, Virginia
| | - Lisa Bradfield
- American Society for Radiation Oncology, Arlington, Virginia
| | - Joseph Califano
- Department of Surgery, University of California San Diego Health, San Diego, California
| | - Jimmy J Caudell
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Christina H Chapman
- Department of Radiation Oncology, Baylor College of Medicine, Houston, Texas
| | - Adam S Garden
- Department of Radiation Oncology, University of Texas - MD Anderson Cancer Center, Houston, Texas
| | - Paul M Harari
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Amanda Helms
- American Society for Radiation Oncology, Arlington, Virginia
| | - Alexander Lin
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ellie Maghami
- Department of Surgery, City of Hope, Duarte, California
| | - Ranee Mehra
- Department of Medical Oncology, University of Maryland Medical School and Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
| | | | - Yelizaveta Shnayder
- Department of Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas
| | - Sharon Spencer
- Department of Radiation Oncology, University of Alabama Heersink School of Medicine, Birmingham, Alabama
| | - Paul L Swiecicki
- Department of Medical Oncology, University of Michigan Rogel Cancer Center, Ann Arbor, Michigan
| | | | - David J Sher
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
90
|
Vasiliadou I, Grose D, Wilson C, Thapa A, Donnelly O, Lee E, Leslie I, Karim M, Hartley A, Partridge S, Medlow K, De Boisanger J, Metcalf R, Williamson A, Haridass A, Noble D, Mactier K, Walter H, Ma N, De Winton E, Cohen J, Rayner L, Geropantas K, Jankowska P, Mason J, Moleron R, Laws K, Ulahannan D, Nallathambi C, Michaelidou A, Nallamilli S, Raouf S, Palmer K, Bienz M, Karet T, Khalique S, Paterson C, Harrington K, Bhide S, Kong A. The use of pembrolizumab monotherapy for the management of head and neck squamous cell carcinoma (HNSCC) in the UK. Int J Cancer 2024; 155:883-893. [PMID: 38685816 DOI: 10.1002/ijc.34963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/01/2024] [Accepted: 01/11/2024] [Indexed: 05/02/2024]
Abstract
Pembrolizumab has received approval in the UK as first-line monotherapy for recurrent and/or metastatic HNSCC (R/M HNSCC) following the results of the KEYNOTE-048 trial, which demonstrated a longer overall survival (OS) in comparison to the EXTREME chemotherapy regimen in patients with a combined positive score (CPS) ≥1. In this article, we provide retrospective real-world data on the role of pembrolizumab monotherapy as first-line systemic therapy for HNSCC across 18 centers in the UK from March 20, 2020 to May 31, 2021. 211 patients were included, and in the efficacy analysis, the objective response rate (ORR) was 24.7%, the median progression-free survival (PFS) was 4.8 months (95% confidence interval [CI]: 3.6-6.1), and the median OS was 10.8 months (95% CI 9.0-12.5). Pembrolizumab monotherapy was well tolerated, with 18 patients having to stop treatment owing to immune-related adverse events (irAEs). 53 patients proceeded to second-line treatment with a median PFS2 of 10.2 months (95% CI: 8.8-11.5). Moreover, patients with documented irAEs had a statistically significant longer median PFS (11.3 vs. 3.3 months; log-rank p value = <.001) and median OS (18.8 vs. 8.9 months; log-rank p value <.001). The efficacy and safety of pembrolizumab first-line monotherapy for HNSCC has been validated using real-world data.
Collapse
Affiliation(s)
- Ifigenia Vasiliadou
- Guy's and St. Thomas NHS Foundation Trust, London, UK
- King's College London, London, UK
| | - Derek Grose
- Beatson West of Scotland Cancer Centre, Glasgow, UK
| | | | - Alekh Thapa
- Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Olly Donnelly
- Portsmouth Hospitals NHS Trust, Portsmouth, Hampshire, UK
| | - Elsa Lee
- Guy's and St. Thomas NHS Foundation Trust, London, UK
- King's College London, London, UK
| | - Isla Leslie
- Royal Marsden NHS Foundation Trust, London, UK
| | | | | | - Sarah Partridge
- Imperial College Healthcare NHS Trust-Charing Cross Hospital, London, UK
| | - Katharine Medlow
- Imperial College Healthcare NHS Trust-Charing Cross Hospital, London, UK
| | - James De Boisanger
- Imperial College Healthcare NHS Trust-Charing Cross Hospital, London, UK
| | | | | | | | | | | | | | - Ning Ma
- University Hospitals of Leicester, Leicester, UK
| | - Emma De Winton
- Royal United Hospitals Bath-NHS Foundation trust, Bath, UK
| | - Jennifer Cohen
- Royal United Hospitals Bath-NHS Foundation trust, Bath, UK
| | - Lindsay Rayner
- Royal United Hospitals Bath-NHS Foundation trust, Bath, UK
| | | | - Petra Jankowska
- Musgrove Park Hospital-Taunton and Somerset NHS Foundation Trust, Taunton, UK
| | - Jessica Mason
- Musgrove Park Hospital-Taunton and Somerset NHS Foundation Trust, Taunton, UK
| | | | - Kirsten Laws
- Aberdeen Royal Infirmary-NHS Grampian, Aberdeen, UK
| | | | | | | | - Susanna Nallamilli
- Maidstone Hospital-Tunbridge Wells Hospital-NHS Trust, Tunbridge Wells, UK
| | - Sherif Raouf
- St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Kieran Palmer
- King's College London, London, UK
- St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | | | | | | | | | - Kevin Harrington
- Royal Marsden NHS Foundation Trust, London, UK
- The Institute of Cancer Research, London, UK
| | - Shreerang Bhide
- Royal Marsden NHS Foundation Trust, London, UK
- The Institute of Cancer Research, London, UK
| | - Anthony Kong
- Guy's and St. Thomas NHS Foundation Trust, London, UK
- King's College London, London, UK
| |
Collapse
|
91
|
Liu C, Li M, Liu X, Shi T, Wang Y, Sui C, Zhang W, Wang B. Evaluating the efficacy and safety of different neoadjuvant immunotherapy combinations in locally advanced HNSCC: a systematic review and meta-analysis. Front Immunol 2024; 15:1467306. [PMID: 39267732 PMCID: PMC11390592 DOI: 10.3389/fimmu.2024.1467306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
Background Immune checkpoint inhibitors have demonstrated promising therapeutic outcomes in recurrent/metastatic (R/M) Head and Neck Squamous Cell Carcinoma (HNSCC), prompting numerous clinical trials to investigate the safety and efficacy of this approach in neoadjuvant therapy. This systematic review aims to consolidate and analyze the findings from various clinical trials combining neoadjuvant immunotherapy for HNSCC, with the goal of identifying the most effective neoadjuvant immunotherapy regimen. Methods The system conducted searches across electronic databases including PubMed, Embase, the Cochrane Library and Web of science from their inception to July 1, 2024. The primary focus was on evaluating efficacy (particularly pathological complete response (pCR), major pathological response (MPR), and overall response rate (ORR)) and safety (primarily assessed by grade 3-4 treatment-related adverse reactions). Results A total of 1943 patients from 32 studies were analyzed. Combining neoadjuvant immunotherapy with chemotherapy or radiotherapy demonstrated superiority over neoadjuvant immunotherapy alone in terms of the MPR rate, while showing no statistically significant difference in the pCR rate. Furthermore, the combination of neoadjuvant immunotherapy with chemotherapy or radiotherapy exhibited a lower CR rate compared to neoadjuvant immunotherapy with radiotherapy alone, but a higher PR rate and SD rate. Apart from the neoadjuvant immunotherapy group in isolation, there were no statistically significant differences in grade ≥3 treatment-related adverse events (TRAEs) and immune-related adverse events (irAEs) among the other three combination therapy groups. Conclusion This systematic review and meta-analysis indicate that patients with locally advanced HNSCC might benefit from neoadjuvant immunotherapy, particularly when used in conjunction with chemotherapy or radiotherapy. Nonetheless, additional data is required to definitively confirm its efficacy. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=553753, identifier CRD42024553753.
Collapse
Affiliation(s)
- Chang Liu
- Department of Burns and Plastic Surgery, Yantaishan Hospital, Yantai, China
| | - Mingzhu Li
- Department of implantology, Affiliated Hospital of Binzhou Medical College, Yantai Stomatology Hospital, Yantai, China
| | - Xiaojie Liu
- Department of Burns and Plastic Surgery, Yantaishan Hospital, Yantai, China
| | - Ting Shi
- Department of Burns and Plastic Surgery, Yantaishan Hospital, Yantai, China
| | - Yun Wang
- Department of Burns and Plastic Surgery, Yantaishan Hospital, Yantai, China
| | - Chaoyang Sui
- Department of Burns and Plastic Surgery, Yantaishan Hospital, Yantai, China
| | - Wenan Zhang
- Department of Burns and Plastic Surgery, Yantaishan Hospital, Yantai, China
| | - Bowen Wang
- Department of Burns and Plastic Surgery, Yantaishan Hospital, Yantai, China
| |
Collapse
|
92
|
Dong S, Zhao M, Zhu J, Li T, Yan M, Xing K, Liu P, Yu S, Ma J, He H. Natural killer cells: a future star for immunotherapy of head and neck squamous cell carcinoma. Front Immunol 2024; 15:1442673. [PMID: 39234249 PMCID: PMC11371580 DOI: 10.3389/fimmu.2024.1442673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 07/15/2024] [Indexed: 09/06/2024] Open
Abstract
The interplay between immune components and the epithelium plays a crucial role in the development and progression of head and neck squamous cell carcinoma (HNSCC). Natural killer (NK) cells, one of the main tumor-killing immune cell populations, have received increasing attention in HNSCC immunotherapy. In this review, we explore the mechanism underlying the interplay between NK cells and HNSCC. A series of immune evasion strategies utilized by cancer cells restrict HNSCC infiltration of NK cells. Overcoming these limitations can fully exploit the antineoplastic potential of NK cells. We also investigated the tumor-killing efficacy of NK cell-based immunotherapies, immunotherapeutic strategies, and new results from clinical trials. Notably, cetuximab, the most essential component of NK cell-based immunotherapy, inhibits the epidermal growth factor receptor (EGFR) signaling pathway and activates the immune system in conjunction with NK cells, inducing innate effector functions and improving patient prognosis. In addition, we compiled information on other areas for the improvement of patient prognosis using anti-EGFR receptor-based monoclonal antibody drugs and the underlying mechanisms and prognoses of new immunotherapeutic strategies for the treatment of HNSCC.
Collapse
Affiliation(s)
- Shuyan Dong
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ming Zhao
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jin Zhu
- Department of Pathology, Xi’an Daxing Hospital, Xi’an, China
| | - Ting Li
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Mingze Yan
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Kaixun Xing
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Peng Liu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Shan Yu
- Department of Pathology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jian Ma
- Department of General Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Immunology, Harbin Medical University, Harbin, China
| | - Hongjiang He
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
93
|
Patin EC, Nenclares P, Chan Wah Hak C, Dillon MT, Patrikeev A, McLaughlin M, Grove L, Foo S, Soliman H, Barata JP, Marsden J, Baldock H, Gkantalis J, Roulstone V, Kyula J, Burley A, Hubbard L, Pedersen M, Smith SA, Clancy-Thompson E, Melcher AA, Ono M, Rullan A, Harrington KJ. Sculpting the tumour microenvironment by combining radiotherapy and ATR inhibition for curative-intent adjuvant immunotherapy. Nat Commun 2024; 15:6923. [PMID: 39134540 PMCID: PMC11319479 DOI: 10.1038/s41467-024-51236-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
The combination of radiotherapy/chemoradiotherapy and immune checkpoint blockade can result in poor outcomes in patients with locally advanced head and neck squamous cell carcinoma (HNSCC). Here, we show that combining ATR inhibition (ATRi) with radiotherapy (RT) increases the frequency of activated NKG2A+PD-1+ T cells in animal models of HNSCC. Compared with the ATRi/RT treatment regimen alone, the addition of simultaneous NKG2A and PD-L1 blockade to ATRi/RT, in the adjuvant, post-radiotherapy setting induces a robust antitumour response driven by higher infiltration and activation of cytotoxic T cells in the tumour microenvironment. The efficacy of this combination relies on CD40/CD40L costimulation and infiltration of activated, proliferating memory CD8+ and CD4+ T cells with persistent or new T cell receptor (TCR) signalling, respectively. We also observe increased richness in the TCR repertoire and emergence of numerous and large TCR clonotypes that cluster based on antigen specificity in response to NKG2A/PD-L1/ATRi/RT. Collectively, our data point towards potential combination approaches for the treatment of HNSCC.
Collapse
Affiliation(s)
- Emmanuel C Patin
- Targeted Therapy Team, The Institute of Cancer Research, London, UK.
| | - Pablo Nenclares
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Charleen Chan Wah Hak
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Magnus T Dillon
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Anton Patrikeev
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | - Lorna Grove
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Shane Foo
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | | | | | - Holly Baldock
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Jim Gkantalis
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | - Joan Kyula
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Amy Burley
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Lisa Hubbard
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Malin Pedersen
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | | | - Alan A Melcher
- Translational Immunotherapy Team, The Institute of Cancer Research, London, UK
| | - Masahiro Ono
- Department of Life Sciences, Imperial College London, London, UK
| | - Antonio Rullan
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Kevin J Harrington
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| |
Collapse
|
94
|
Guo LF, Rao MY, Yu YF, Lin Q, Wu SG. The addition of nimotuzumab during concurrent chemoradiotherapy improved survival outcomes in locally advanced nasopharyngeal carcinoma patients with optimal response to induction chemotherapy. BMC Cancer 2024; 24:950. [PMID: 39095737 PMCID: PMC11297786 DOI: 10.1186/s12885-024-12731-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024] Open
Abstract
OBJECTIVE To investigate the impact of response to induction chemotherapy (IC) on survival outcomes in patients with locally advanced nasopharyngeal carcinoma (LANPC) and evaluate the efficacy of adding nimotuzumab to concurrent chemoradiotherapy (CCRT) based on different responses to IC. METHODS We retrospectively included patients with stage III-IVA NPC who underwent IC with and without nimotuzumab during CCRT. Statistical analysis included the chi-square test, propensity score matching, Kaplan-Meier survival analysis, and Cox proportional hazards model. RESULTS Among 383 identified patients, 216 (56.4%) received nimotuzumab during CCRT, while 167 (43.6%) did not. Following IC, 269 (70.2%) patients showed a complete response (CR) or partial response (PR), and 114 (29.8%) had stable disease (SD) or progressive disease (PD). The response to IC independently influenced disease-free survival (DFS) and overall survival (OS). Patients achieving CR/PR demonstrated significantly higher 3-year DFS (80.3% vs. 70.6%, P = 0.031) and OS (90.9% vs. 83.2%, P = 0.038) than those with SD/PD. The addition of nimotuzumab during CCRT significantly improved DFS (P = 0.006) and OS (P = 0.037) for CR/PR patients but not for those with SD/PD. CONCLUSIONS This study emphasizes the importance of IC response in LANPC and highlights the potential benefits of nimotuzumab during CCRT for improving survival outcomes in CR/PR patients. Tailored treatment approaches for SD/PD patients warrant further investigation.
Collapse
Affiliation(s)
- Lin-Feng Guo
- Department of Radiation Oncology, Xiamen Cancer Quality Control Center, Xiamen Key Laboratory of Radiation Oncology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, People's Republic of China
| | - Ming-Yue Rao
- Department of Radiology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, People's Republic of China
| | - Yi-Feng Yu
- Department of Radiation Oncology, Xiamen Cancer Quality Control Center, Xiamen Key Laboratory of Radiation Oncology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, People's Republic of China
| | - Qin Lin
- Department of Radiation Oncology, Xiamen Cancer Quality Control Center, Xiamen Key Laboratory of Radiation Oncology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, People's Republic of China.
| | - San-Gang Wu
- Department of Radiation Oncology, Xiamen Cancer Quality Control Center, Xiamen Key Laboratory of Radiation Oncology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, People's Republic of China.
| |
Collapse
|
95
|
Lynch C, Pitroda SP, Weichselbaum RR. Radiotherapy, immunity, and immune checkpoint inhibitors. Lancet Oncol 2024; 25:e352-e362. [PMID: 39089313 DOI: 10.1016/s1470-2045(24)00075-5] [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: 12/12/2023] [Revised: 01/11/2024] [Accepted: 01/29/2024] [Indexed: 08/03/2024]
Abstract
Radiotherapy exerts immunostimulatory and immunosuppressive effects, both locally, within the irradiated tumour microenvironment, and systemically, outside the radiation field. Inspired by preclinical data that showed synergy between radiotherapy and immune checkpoint inhibitors, multiple clinical trials were initiated with the hypothesis that combined treatment with radiotherapy and immune checkpoint inhibitors could stimulate a robust systemic immune response and improve clinical outcomes. However, despite early optimism, radioimmunotherapy trials in the curative and metastatic settings have met with little success. In this Review, we summarise the immunostimulatory effects of radiotherapy that provided the theoretical basis for trials of combination radiotherapy and immune checkpoint inhibitors. We also discuss findings from clinical trials incorporating radiotherapy and immune checkpoint inhibitors and examine the success of these trials in the context of the immunosuppressive effects of radiotherapy. We conclude by highlighting targets for relieving radiotherapy-induced immunosuppression with the goal of enhancing the combined effects of radiotherapy and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Connor Lynch
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Sean P Pitroda
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
96
|
Wu C, Kuzmin P, Julian R. De-Escalation Strategies in HPV-Associated Oropharynx Cancer: A Historical Perspective with Future Direction. Cancers (Basel) 2024; 16:2733. [PMID: 39123461 PMCID: PMC11311653 DOI: 10.3390/cancers16152733] [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: 06/03/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
The incidence of HPV-related oropharyngeal cancers has increased in recent decades. While cure rates exceed those of HPV-negative head and neck cancers, both acute and long-term sequelae of chemotherapy, radiation and surgery have led to clinical investigation into de-escalation of treatment. De-escalation trials have sought to reduce long-term treatment-related morbidity by altering or omitting chemotherapy, reducing radiation, or incorporating less invasive surgical resection through transoral surgery. More recent approaches include the use of novel agents such as immunotherapy in place of cisplatin. With the advent of tumor-tissue-modified HPV DNA detection and monitoring in blood, new strategies incorporating this biomarker are being developed.
Collapse
Affiliation(s)
- Clinton Wu
- Department of Internal Medicine, University of Arizona, Tucson, AZ 85719, USA
| | - Paulina Kuzmin
- Department of Internal Medicine, University of Arizona, Tucson, AZ 85719, USA
| | - Ricklie Julian
- Department of Internal Medicine, University of Arizona, Tucson, AZ 85719, USA
- Department of Hematology and Oncology, University of Arizona, Tucson, AZ 85719, USA
| |
Collapse
|
97
|
Okano S. Immunotherapy for head and neck cancer: Fundamentals and therapeutic development. Auris Nasus Larynx 2024; 51:684-695. [PMID: 38729034 DOI: 10.1016/j.anl.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/03/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024]
Abstract
Squamous cell carcinoma of the head and neck (SCCHN) has been treated by multidisciplinary therapy consisting of surgery, radiotherapy, and cancer chemotherapy, but the recent advent of immunotherapy has produced significant changes in treatment systems and the results of these therapies. Immunotherapy has greatly improved the outcome of recurrent metastatic SCCHN, and the development of new treatment methods based on immunotherapy is now being applied not only to recurrent metastatic cases but also to locally advanced cases. To understand and practice cancer immunotherapy, it is important to understand the immune environment surrounding cancer, and the changes to which it is subject. Currently, the anti-PD-1 antibody drugs nivolumab and pembrolizumab are the only immunotherapies with proven efficacy in head and neck cancer. However, anti-PD-L1 and anti-CTLA-4 antibody drugs have also been shown to be useful in other types of cancer and are being incorporated into clinical practice. In head and neck cancer, numerous clinical trials have aimed to improve efficacy and safety by combining immunotherapy with other drug therapies and treatment modalities. Combinations of immunotherapy with cancer drugs with different mechanisms of action (cytotoxic agents, molecular-targeted agents, immune checkpoint inhibitors), as well as with radiation therapy and surgery are being investigated, and have the potential to significantly change medical care for these patients. The application of cancer immunotherapy not only to daily clinical practice but also to further therapeutic development requires a clear and complete understanding of the fundamentals of cancer immunotherapy, and knowledge of the numerous clinical studies conducted, both past and present. The results of these trials are numerous, both positive and negative, and a comprehensive understanding of this wide range of completed and ongoing clinical trials is critical to a systematic and comprehensive understanding of their scope and lessons learnt. In this article, after outlining the concepts of ``cancer immune cycle,'' ``cancer immune editing,'' and ``tumor microenvironment'' to provide an understanding of the basics of cancer immunity, we summarize the basics and clinical trial data on representative immune checkpoint inhibitors used in various cancer types, as well as recent therapeutic developments in cancer immunotherapy and the current status of these new treatments.
Collapse
Affiliation(s)
- Susumu Okano
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577, Japan.
| |
Collapse
|
98
|
Wu X, Wu J. A polo-like kinase 1 inhibitor enhances erastin sensitivity in head and neck squamous cell carcinoma cells in vitro. Cancer Chemother Pharmacol 2024; 94:183-195. [PMID: 38536443 PMCID: PMC11390781 DOI: 10.1007/s00280-024-04654-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/14/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND Polo-like kinase 1 (PLK1) is a critical therapeutic target in the treatment of head and neck squamous cell carcinoma (HNSCC). The objective of this study was to investigate the therapeutic effect of the combination of BI 2536, a PLK1 inhibitor, and erastin, a ferroptosis inducer, in HNSCC. METHODS The proliferation, invasion, and migration abilities of Tu177 and FaDu cells upon exposure to BI 2536 and erastin, used in combination or alone, were tested. Fe2+, glutathione (GSH), and malondialdehyde (MDA) detection kits were used to determine whether the addition of BI 2536 enhanced the accumulation of Fe2+ and MDA, along with the depletion of GSH. Quantitative real-time PCR, western blot analyses were performed to investigate whether BI 2536 further altered the mRNA and expression level of ferroptosis genes. Furthermore, si PLK1 was used to investigate whether targeting PLK1 gene promoted erastin-induced ferroptosis. RESULTS The combination of BI 2536 and erastin exerted a stronger cytotoxicity than treatment with a single agent. Compared with erastin treatment alone, the combination of BI 2536 and erastin lowered the ability of tumor cells to self-clone, invade, and migrate. BI 2536 enhanced the accumulation of Fe2+ and MDA, and the depletion of GSH. BI 2536 increased erastin-induced changes in ferroptosis-related gene mRNA and expression. Importantly, targeting PKL1 enhanced the anti-cancer effect of erastin. CONCLUSION BI 2536 enhanced the sensitivity of HNSCC cells to erastin, which provides a new perspective for cancer treatment.
Collapse
Affiliation(s)
- Xiangping Wu
- Department of Otolaryngology-Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Jing Wu
- Department of Otolaryngology-Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
| |
Collapse
|
99
|
Kerr CP, Sheehan-Klenk J, Grudzinski JJ, Adam DP, Nguyen TPT, Ferreira CA, Bates AM, Jin WJ, Kwon O, Olson AP, Lin W, Hyun M, Jagodinsky JC, Powers M, Sriramaneni RN, Clark PA, Shea AG, Rojas HC, Choi C, Massey CF, Zangl LM, Pinchuk AN, Aluicio-Sarduy E, Kim K, Engle JW, Hernandez R, Bednarz BP, Weichert JP, Morris ZS. Effects of clinically relevant radionuclides on the activation of a type I interferon response by radiopharmaceuticals in syngeneic murine tumor models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602990. [PMID: 39071353 PMCID: PMC11275738 DOI: 10.1101/2024.07.10.602990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Radiopharmaceutical therapies (RPT) activate a type I interferon (IFN1) response in tumor cells. We hypothesized that the timing and amplitude of this response varies by isotope. We compared equal doses delivered by 90 Y, 177 Lu, and 225 Ac in vitro as unbound radionuclides and in vivo when chelated to NM600, a tumor-selective alkylphosphocholine. Response in murine MOC2 head and neck carcinoma and B78 melanoma was evaluated by qPCR and flow cytometry. Therapeutic response to 225 Ac-NM600+anti-CTLA4+anti-PD-L1 immune checkpoint inhibition (ICI) was evaluated in wild-type and stimulator of interferon genes knockout (STING KO) B78. The timing and magnitude of IFN1 response correlated with radionuclide half-life and linear energy transfer. CD8 + /Treg ratios increased in tumors 7 days after 90 Y- and 177 Lu-NM600 and day 21 after 225 Ac-NM600. 225 Ac-NM600+ICI improved survival in mice with WT but not with STING KO tumors, relative to monotherapies. Immunomodulatory effects of RPT vary with radioisotope and promote STING-dependent enhanced response to ICIs in murine models. Teaser This study describes the time course and nature of tumor immunomodulation by radiopharmaceuticals with differing physical properties.
Collapse
|
100
|
Tian Y, Yin Z, Zhang C, Li Z, Wang Y, Zhang K, Chen F, Dang Q. Differences in the risk of immune-related pneumonitis between PD-1 and PD-L1 inhibitors: a meta-analysis according to the new mirror-principle and PRISMA guidelines. Cancer Immunol Immunother 2024; 73:162. [PMID: 38953977 PMCID: PMC11219650 DOI: 10.1007/s00262-024-03736-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 05/15/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE To compare the risk of immune-associated pneumonitis between PD-1 and PD-L1 inhibitors, the meta-analysis was designed. METHOD The difference in risk of immune-associated pneumonitis between PD-1 and PD-L1 inhibitors was assessed by two different meta-analysis methods, the Mirror-pairing and the PRISMA guidelines. RESULTS A total of eighty-eight reports were used for meta-analysis, while thirty-two studies were used for the Mirror-pairing. Both PD-1 and PD-L1 inhibitors (used alone or combined with chemotherapy) increased the risk of developing immune-related pneumonitis (P < 0.00001; P < 0.00001). Based on indirect analyses results (subgroup analyses), the risk of PD-L1-induced pneumonitis was weaker than that of PD-1 inhibitors when the control group was chemotherapy (OR = 3.33 vs. 5.43) or placebo (OR = 2.53 vs. 3.19), while no obvious significant differences were found (P = 0.17; P = 0.53). For the Mirror-pairing-based meta-analysis, the risk of PD-1-induced pneumonitis was significantly higher than that of PD-L1 inhibitors (OR = 1.46, 95%CI [1.08, 1.98], I2 = 0%, Z = 2.47 (P = 0.01)). However, this difference was not significant, when they were combined with chemotherapy (OR = 1.05, 95%CI [0.68, 1.60], I2 = 38%, Z = 0.21 (P = 0.84)). CONCLUSION Both PD-1 and PD-L1 inhibitors increased the risk of immune-related pneumonitis, while the risk of PD-1-induced pneumonitis was significantly higher than that of PD-L1 inhibitors.
Collapse
Affiliation(s)
- Yuan Tian
- Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Huaiyin District, Jinan City, 250117, Shandong, People's Republic of China
- Radiotherapy Department, Shandong Second Provincial General Hospital, Shandong University, Jinan, 250299, Shandong, People's Republic of China
| | - Zongxiu Yin
- Department of Pulmonary and Critical Care Medicine, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, Shandong, People's Republic of China
| | - Chi Zhang
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, People's Republic of China
| | - Zhuoqi Li
- Radiotherapy Department, Shandong Second Provincial General Hospital, Shandong University, Jinan, 250299, Shandong, People's Republic of China
| | - Yuanyuan Wang
- Department of Oncology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250299, Shandong, People's Republic of China
| | - Kai Zhang
- General Surgery Department, Wen-Shang County People's Hospital, Wenshang, 272500, Shandong, People's Republic of China
| | - Feng Chen
- Department of Thoracic Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, People's Republic of China
| | - Qi Dang
- Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Huaiyin District, Jinan City, 250117, Shandong, People's Republic of China.
| |
Collapse
|