1
|
Rajendran S, Surabhi RP, Kumar AS, Gopinath P, Kanakaveti V, Shanmugasundaram G, Michael Gromiha M, Rayala SK, Venkatraman G. P21-Activated Kinase 1 (PAK1) Modulates Therapeutic Response to Ionizing Radiation in Head and Neck Squamous Cell Carcinoma Cells. Mol Carcinog 2025; 64:970-984. [PMID: 40099538 DOI: 10.1002/mc.23902] [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: 08/26/2024] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
Head and neck squamous cell carcinoma (HNSCC) continues to be a formidable epithelial malignancy characterized by late-stage detection and recurrence impacting survival. P21-activated kinase-1 (PAK1) was reported to be overexpressed in head and neck cancers and activated by ionizing radiation (IR), affecting treatment outcomes. Present investigations revealed that PAK1 silencing on HNSCC cells reverted the aggressive phenotype and showed impaired DNA damage repair upon IR exposure. Further HNSCC cells were resistant to IR up to 30 Gy with elevated pPAK1 levels. Radiation-resistant (RR) HNSCC cells expressed radiation-resistant markers, namely MRE-11 and NME-1; stemness markers-OCT4 and SOX2; and EMT & metastasis markers-vimentin, snail, and α-smooth muscle actin (α-SMA). In addition, HNSCC RR cells showed increased levels of DNA damage response protein H2AX, indicative of an aggressive phenotype with an augmented DNA repair machinery and a potential target for inhibition. Since H2AX appears to be a mechanistic hub for PAK1-induced radiation resistance, using in silico methods, peptides were designed, and the PL-8 peptide was chosen to target the phosphorylation of H2AX, which could enhance the sensitivity to IR and push the cells to radiation-induced cell death. PL-8 peptide inhibited H2AX phosphorylation on HNSCC cells and triggered radiation-induced cell death as determined by functional assays. The present study reveals PAK1 induced in HNSCC cells by IR and causes resistance by enhancing DNA damage response mediated through γH2AX. To counteract this complex molecular interplay, we propose inhibiting γH2AX formation & silencing PAK1 appears to be a probable way forward in HNSCC.
Collapse
Affiliation(s)
- Swetha Rajendran
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, Tamil Nadu, India
| | - Rohan Prasad Surabhi
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, Tamil Nadu, India
| | - A Satheesh Kumar
- Department of Biotechnology, Indian Institute of Technology Madras, Guindy, Chennai, Tamil Nadu, India
| | - Prarthana Gopinath
- Department of Biotechnology, Indian Institute of Technology Madras, Guindy, Chennai, Tamil Nadu, India
| | - Vishnupriya Kanakaveti
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, California, USA
| | | | - M Michael Gromiha
- Department of Biotechnology, Indian Institute of Technology Madras, Guindy, Chennai, Tamil Nadu, India
| | - Suresh Kumar Rayala
- Department of Biotechnology, Indian Institute of Technology Madras, Guindy, Chennai, Tamil Nadu, India
| | - Ganesh Venkatraman
- Department of Bio-Medical Sciences, School of Bio Sciences & Technology, Vellore Institute of Technology Vellore, Vellore, Tamil Nadu, India
| |
Collapse
|
2
|
Hsieh JCH, Lien MY, Chang PH, Wang HM, Yeh KY, Ho CL, Hsieh CY, Hsieh MC, Chen JH. UFUR maintenance therapy significantly improves survival in locally advanced head and neck squamous cell carcinoma following definitive chemoradiotherapy. Cancer Cell Int 2025; 25:187. [PMID: 40410757 PMCID: PMC12102819 DOI: 10.1186/s12935-025-03807-w] [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: 10/31/2024] [Accepted: 05/04/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) presents significant therapeutic challenges, particularly in patients with advanced disease. Despite advancements in treatment, high recurrence rates and poor overall survival (OS) remain major concerns. This study evaluates the impact of tegafur-uracil (UFUR) maintenance therapy on survival outcomes in patients with advanced HNSCC following definitive chemoradiotherapy. METHODS A cohort of 424 advanced HNSCC patients treated with definitive chemoradiotherapy were analyzed, with a median follow-up of 25 months. Patients were stratified into UFUR (+) and UFUR (-) groups, with baseline characteristics balanced across both arms. Oncologic outcomes, including recurrence-free survival (RFS), OS, locoregional recurrence-free survival (LRFS), and distant metastasis-free survival (DMFS) were compared between these groups. RESULTS UFUR maintenance therapy significantly reduced recurrence rates (34% vs. 47%, p < 0.002), driven primarily by improving locoregional control (23% vs. 37%, p < 0.001). While distant metastasis rates were similar between groups, UFUR (+) patients demonstrated a markedly improved median OS (51.6 months vs. 24.3 months, p < 0.001). The UFUR (+) group also showed superior median LRFS (36.9 months vs. 20.2 months, p = 0.003) and DMFS (44.0 months vs. 23.5 months, p = 0.010). Subgroup analysis confirmed the benefits of UFUR maintenance across different stages of disease. Multivariate analysis identified UFUR maintenance, gender, and T stage as independent predictors of survival. CONCLUSION UFUR maintenance therapy significantly improves survival outcomes in patients with advanced HNSCC following definitive chemoradiotherapy, particularly through enhanced locoregional control, and should be considered a key component of personalized treatment strategies.
Collapse
Affiliation(s)
- Jason Chia-Hsun Hsieh
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan
- Division of Hematology-Oncology, Department of Internal Medicine, New Taipei City Municipal TuCheng Hospital, New Taipei City, 236, Taiwan
| | - Ming-Yu Lien
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Pei-Hung Chang
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Keelung, Keelung City, 204, Taiwan
| | - Hung-Ming Wang
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan
| | - Kun-Yun Yeh
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Keelung, Keelung City, 204, Taiwan
| | - Ching-Liang Ho
- Division of Hematology and Oncology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, No.325, Sec. 2, Chenggong Rd., Neihu District, Taipei City, 11490, Taiwan
- Division of Hematology and Oncology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Ching-Yun Hsieh
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Meng-Che Hsieh
- Department of Hematology-Oncology, E-Da Cancer Hospital, No.21, E-Da Rd., Yan-Chao Dist, Kaohsiung, 807, Taiwan.
- College of Medicine, I-Shou University, Kaohsiung, 824, Taiwan.
| | - Jia-Hong Chen
- Division of Hematology and Oncology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, No.325, Sec. 2, Chenggong Rd., Neihu District, Taipei City, 11490, Taiwan.
| |
Collapse
|
3
|
Hildebrand LS, Jost T, Schindler M, Derer A, Fuhrmann G, Fietkau R, Distel LV. Inhibiting NHEJ in HNSCC cell lines by the ligase IV inhibitor SCR130 has limited radiosensitizing effects. Sci Rep 2025; 15:17871. [PMID: 40404928 PMCID: PMC12098888 DOI: 10.1038/s41598-025-03159-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025] Open
Abstract
Radiotherapy (RT) is a relevant treatment for head and neck squamous cell carcinoma (HNSCC) patients but radioresistance, which depends on DNA damage response (DDR), restrains outcome. Therefore, manipulating DDR by small molecule inhibitors (SMI) is a promising treatment option. The main DNA double strand break (DSB) repair mechanisms in healthy mammalian cells are homologous recombination (HR) and non-homologous end joining (NHEJ). It is known that HR is already often impaired in tumors because of cancerous transitions. Therefore, additionally inhibiting NHEJ is a possibility to specifically target tumor cells and spare healthy tissue, which has the alternative DSB repair mechanism available. We treated HNSCC and healthy fibroblast cell lines with 30 µM of the ligase IV inhibitor SCR130 and a single dose of 2 Gy (Gy) ionizing radiation (IR) to investigate the inhibitor's radiosensitizing effect. In short, the effect of SCR130 in combination with IR on cell death, clonogenicity, and DNA damage is limited and highly cell line specific. Nevertheless, SCR130 increases the number of cells in G0/G1 phase concomitant with gained p21 expression consistently. We suggest that SCR130 in combination with IR has anti-proliferative effects, but an escape of the cells by upregulation of ligase IV resulting from the treatment is possible.
Collapse
Affiliation(s)
- Laura S Hildebrand
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Tina Jost
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Department of Radiation Oncology, Translational Radiobiology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany
| | - Marion Schindler
- Department of Biology, Pharmaceutical Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Staudtstraße 5, 91058, Erlangen, Germany
| | - Anja Derer
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Department of Radiation Oncology, Translational Radiobiology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany
| | - Gregor Fuhrmann
- Department of Biology, Pharmaceutical Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Staudtstraße 5, 91058, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Luitpold V Distel
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany.
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany.
| |
Collapse
|
4
|
Chen Y, Zhang Z, Yin R, Li Q, Zhang W. Bibliometric analysis of research trends on the combination of radiotherapy and PARP inhibitors in solid tumors. Front Pharmacol 2025; 16:1603573. [PMID: 40421223 PMCID: PMC12104260 DOI: 10.3389/fphar.2025.1603573] [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: 03/31/2025] [Accepted: 04/29/2025] [Indexed: 05/28/2025] Open
Abstract
Introduction Radiotherapy has served as a cornerstone in cancer treatment for over a century. However, the efficacy of radiotherapy is often compromised by the intrinsic and acquired radioresistance of tumors, which can lead to treatment failure and disease recurrence. Recent advancements in preclinical and clinical research have highlighted the potential synergistic efficacy of combining radiotherapy with poly-ADP-ribose polymerase inhibitors (PARPi), offering promising therapeutic avenues for solid tumors. This study employs bibliometric analysis to systematically evaluate the evolution, trends, and intellectual landscape of research on the combination of radiotherapy and PARPi in solid tumors. Methods Publications addressing the combination of radiotherapy and PARPi for solid tumors between 2005 and 2024 were retrieved from the Web of Science Core Collection (WOSCC) database. Bibliometric assessments were conducted using VOSviewer and CiteSpace to analyze publication trends, collaborative networks, and research foci. Results A total of 901 articles were included. The United States dominated research output, with the University of Texas MD Anderson Cancer Center identified as the most productive institution. Hannah Farmer emerged as the most frequently cited author. Keywords co-occurrence analysis revealed a thematic shift from foundational studies on molecular mechanisms, such as DNA damage response and mechanism of action of PARPi, toward clinical investigations evaluating combination therapy efficacy and safety in trials. Conclusion This bibliometric analysis underscores the rapid growth of research on radiotherapy and PARPi combination therapy, with the United States maintaining a leading role due to its extensive scientific infrastructure and collaborative networks. The field has transitioned from mechanistic explorations to translational and clinical applications, reflecting progress toward therapeutic optimization. These findings provide a comprehensive overview of the knowledge structure within this domain and serve as a strategic reference for guiding future research priorities and clinical implementations.
Collapse
Affiliation(s)
- Yuxin Chen
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Zhengkun Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan, China
- College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Rutie Yin
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Qingli Li
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Wenhao Zhang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
5
|
Pang G, Lu Y, Xu Y, Meng Q, Song J. The investigation of opening modes of head and neck thermoplastic mask for radiotherapy based on finite element analysis. Radiat Oncol 2025; 20:64. [PMID: 40301880 PMCID: PMC12038929 DOI: 10.1186/s13014-025-02648-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 04/24/2025] [Indexed: 05/01/2025] Open
Abstract
Radiotherapy is a crucial treatment modality for head and neck tumors. Precise patient positioning is vital for ensuring the reproducibility and accuracy of the treatment. Clinically, thermoplastic head and neck masks are used for positioning, and patients often require openings in these masks. However, the relationship between opening locations and the fixation effectiveness of the masks has not been thoroughly studied. This study reconstructs a patient's imaging data to create finite element models with 10 different opening patterns. By assigning various thicknesses and material properties to the fixation masks, we analyzed the displacement distribution of different opening models under diverse loading conditions. The results indicate that the opening location significantly impacts fixation effectiveness under different loading conditions. In particular, the opening in the forehead area has the most significant impact on the fixation effect of the mask near the region of interest (ROI). Therefore, in clinical practice, the design of openings in the forehead area should be carefully considered. This study provides valuable insights into the fixation effectiveness of thermoplastic masks and serves as a reference for future personalized positioning treatments.
Collapse
Affiliation(s)
- Guobao Pang
- Cancer Center, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Ying Lu
- Cancer Center, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Provincial Key Laboratory for Translational Nuclear Medicine and Precision Protection, Taiyuan, China
| | - Yannan Xu
- Cancer Center, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Qiaoyu Meng
- Institute of Advanced Structure Technology, Beijing Institute of Technology, Beijing, China.
| | - Jianbo Song
- Cancer Center, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
6
|
Kaczmarczyk O, Augustyniak D, Żak A. Imaging of Hydrated and Living Cells in Transmission Electron Microscope: Summary, Challenges, and Perspectives. ACS NANO 2025; 19:12710-12733. [PMID: 40156542 PMCID: PMC11984313 DOI: 10.1021/acsnano.5c00871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 04/01/2025]
Abstract
Transmission electron microscopy (TEM) is well-known for performing in situ studies in the nanoscale. Hence, scientists took this opportunity to explore the subtle processes occurring in living organisms. Nevertheless, such observations are complex─they require delicate samples kept in the liquid phase, low electron dose, and proper cell viability verification methods. Despite being highly demanding, so-called "live-cell" experiments have seen some degree of success. The presented review consists of an exhaustive literature review on reported "live-cell" studies and associated subjects, including liquid phase imaging, electron radiation interactions with liquids, and methods for cell viability testing. The challenges of modern, reliable research on living organisms are widely explained and discussed, and future perspectives for developing these techniques are presented.
Collapse
Affiliation(s)
- Olga Kaczmarczyk
- Institute
of Advanced Materials, Wroclaw University
of Science and Technology, 50-370 Wroclaw, Poland
| | - Daria Augustyniak
- Department
of Pathogen Biology and Immunology, Faculty of Biological Sciences, University of Wroclaw, 51-148 Wroclaw, Poland
| | - Andrzej Żak
- Institute
of Advanced Materials, Wroclaw University
of Science and Technology, 50-370 Wroclaw, Poland
- Department
of Material Science and Engineering, Massachusetts
Institute of Science and Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
7
|
Anand P, Chhimwal J, Dhiman S, Yamini, Patial V, Das P, Ahmed Z, Nandi U, Tavassoli M, Padwad Y. Evaluation of Pyrrolone-Fused Benzosuberene MK2 Inhibitors as Promising Therapeutic Agents for HNSCC: In Vitro Efficacy, In-Vivo Safety, and Pharmacokinetic Profiling. Drug Dev Res 2025; 86:e70062. [PMID: 40009048 DOI: 10.1002/ddr.70062] [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: 08/02/2024] [Revised: 10/18/2024] [Accepted: 01/17/2025] [Indexed: 02/27/2025]
Abstract
MAPKAPK2/MK2 is well implicated in the progression of Head and Neck Squamous Cell Carcinoma (HNSCC), and potent MK2-inhibitors are required to suppress its activity. Several MK2-inhibitors have been developed in recent years to combat its effects on cancer. However, inadequate solubility, insufficient cellular permeability, systemic toxicity-mediated side effects, and low bioavailability have severely impeded the advancement of MK2-inhibitors to clinical trials. This void necessitates research to develop less toxic and more bioavailable potent MK2-inhibitors in HNSCC. In the present article, we have evaluated the in-vitro efficacy, in-vivo single-dose acute toxicity, and in-vivo pharmacokinetic profiling of recently developed PfBS (pyrrolone-fused benzosuberene) MK2-inhibitor analogues against HNSCC. The PfBS MK2 inhibitor analogues impeded HPV+ and HPV- HNSCC cell proliferation and two-dimensional migration. Moreover, MK2-inhibitors lowered HNSCC cell clonogenic survival in a dose-dependent manner, significantly enhancing radiation-induced cell death via exerting radio-sensitization effects. Furthermore, γ-H2AX immunostaining revealed that PfBS analogues impaired DNA damage repair in HNSCC cells exposed to gamma radiation. In mice, PfBS MK2 inhibitors at 300 mg/kg were well-tolerated without any lethal effects. Pharmacokinetic studies showed that PfBS analogues exhibited rapid absorption (Tmax), adequate plasma concentration above the micromolar level (C0 or Cmax), limited tissue distribution (Vd), and faster elimination from the body (Cl). Overall, this study summarizes in-vitro efficacy, safety, and pharmacokinetics of developed MK2-inhibitors and opens doors for pharmacodynamics and mechanism of action study of most effective leads in HNSCC.
Collapse
Affiliation(s)
- Prince Anand
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, Himachal Pradesh, India
- Centre for Host-Microbiome Interactions, King's College London, London, London, United Kingdom
- Academy of Scientific and Innovative Research, AcSIR, Ghaziabad, Uttar Pradesh, India
| | - Jyoti Chhimwal
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, Himachal Pradesh, India
- Academy of Scientific and Innovative Research, AcSIR, Ghaziabad, Uttar Pradesh, India
| | - Sumit Dhiman
- Academy of Scientific and Innovative Research, AcSIR, Ghaziabad, Uttar Pradesh, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Jammu, Jammu & Kashmir, India
| | - Yamini
- Academy of Scientific and Innovative Research, AcSIR, Ghaziabad, Uttar Pradesh, India
- Chemical Technology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, Himachal Pradesh, India
| | - Vikram Patial
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, Himachal Pradesh, India
- Academy of Scientific and Innovative Research, AcSIR, Ghaziabad, Uttar Pradesh, India
| | - Pralay Das
- Academy of Scientific and Innovative Research, AcSIR, Ghaziabad, Uttar Pradesh, India
- Chemical Technology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, Himachal Pradesh, India
| | - Zabeer Ahmed
- Academy of Scientific and Innovative Research, AcSIR, Ghaziabad, Uttar Pradesh, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Jammu, Jammu & Kashmir, India
| | - Utpal Nandi
- Academy of Scientific and Innovative Research, AcSIR, Ghaziabad, Uttar Pradesh, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Jammu, Jammu & Kashmir, India
- Chemical Sciences, Unified Academic Campus, Bose Institute, Kolkata, West Bengal, India
| | - Mahvash Tavassoli
- Centre for Host-Microbiome Interactions, King's College London, London, London, United Kingdom
| | - Yogendra Padwad
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, Himachal Pradesh, India
- Academy of Scientific and Innovative Research, AcSIR, Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
8
|
Guo Z, Li K, Ren X, Wang X, Yang D, Ma S, Zeng X, Zhang P. The role of the tumor microenvironment in HNSCC resistance and targeted therapy. Front Immunol 2025; 16:1554835. [PMID: 40236700 PMCID: PMC11996806 DOI: 10.3389/fimmu.2025.1554835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
The prognosis for head and neck squamous cell carcinoma (HNSCC) remains unfavorable, primarily due to significant therapeutic resistance and the absence effective interventions. A major obstacle in cancer treatment is the persistent resistance of cancer cells to a variety of therapeutic modalities. The tumor microenvironment (TME) which includes encompasses all non-malignant components and their metabolites within the tumor tissue, plays a crucial role in this context. The distinct characteristics of the HNSCC TME facilitate tumor growth, invasion, metastasis, and resistance to treatment. This review provides a comprehensive overview of the HNSCC TME components, with a particular focus on tumor-associated macrophages (TAMs), regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), cancer-associated fibroblasts (CAFs), the extracellular matrix, reprogrammed metabolic processes, and metabolic products. It elucidates their contributions to modulating resistance to chemotherapy, radiotherapy, targeted therapy, and immunotherapy in HNSCC, and explores novel therapeutic strategies targeting the TME for HNSCC management.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Peng Zhang
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| |
Collapse
|
9
|
Sobti A, Skinner H, Wilke CT. Predictors of Radiation Resistance and Novel Radiation Sensitizers in Head and Neck Cancers: Advancing Radiotherapy Efficacy. Semin Radiat Oncol 2025; 35:224-242. [PMID: 40090749 DOI: 10.1016/j.semradonc.2025.02.008] [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: 10/15/2024] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 03/18/2025]
Abstract
Radiation resistance in head and neck squamous cell carcinoma (HNSCC), driven by intrinsic and extrinsic factors, poses a significant challenge in radiation oncology. The key contributors are tumor hypoxia, cancer stem cells, cell cycle checkpoint activation, and DNA repair processes (homologous recombination and non-homologous end-joining). Genetic modifications such as TP53 mutations, KRAS mutations, EGFR overexpression, and abnormalities in DNA repair proteins like BRCA1/2 additionally affect radiation sensitivity. Novel radiosensitizers targeting these pathways demonstrate the potential to overcome resistance. Hypoxia-activated drugs and gold nanoparticles enhance the efficacy of radiotherapy and facilitate targeted distribution. Integrating immunotherapy, especially immune checkpoint inhibitors, with radiation therapy, enhances anti-tumor responses and reduces resistance. Epigenetic alterations, such as DNA methylation and histone acetylation, significantly influence radiation response, with the potential for sensitization through histone deacetylase inhibitors and non-coding RNA regulators. Metabolic changes linked to glucose, lipid, and glutamine metabolism influence radiosensitivity, uncovering new targets for radiosensitization. Human papillomavirus (HPV)-associated malignancies exhibit increased radiosensitivity relative to other tumors due to impaired DNA repair mechanisms and heightened immunogenicity. Furthermore, understanding the interplay between HPV oncoproteins and p53 functionality can enhance treatment strategies for HPV-related cancers. Using DNA damage response inhibitors (PARP, ATM/ATR), cell cycle checkpoint inhibitors (WEE1, CHK1/2), and hypoxia-targeted agents as radiosensitizing strategies exhibit considerable promise. Immunomodulatory approaches, including PD-1 and CTLA-4 inhibitors in conjunction with radiation, enhance anti-tumor immunity. Future directions emphasize personalized radiation therapy using genetics, sophisticated medication delivery systems, adaptive radiotherapy, and real-time monitoring. These integrated strategies seek to diminish radiation resistance and improve therapeutic efficacy in HNSCC.
Collapse
Affiliation(s)
- Aastha Sobti
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA
| | - Heath Skinner
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA
| | - Christopher T Wilke
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA..
| |
Collapse
|
10
|
Špiljak B, Sauerborn D, Tomas M, Gregorić Butina B, Mahovne I, Erić S, Vidaković B, Lešić S. Aggressive Squamoid Eccrine Ductal Carcinoma of the Face: A Rare and Challenging Diagnosis-Case Report and Literature Review. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:612. [PMID: 40282903 PMCID: PMC12028541 DOI: 10.3390/medicina61040612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025]
Abstract
Background: Squamoid eccrine ductal carcinoma (SEDC) is an exceedingly rare and aggressive cutaneous adnexal malignancy, with fewer than 100 reported cases. Its histopathologic overlap with squamous cell carcinoma (SCC) frequently leads to misdiagnosis, delaying appropriate management. Unlike SCC, SEDC exhibits biphasic differentiation, deep infiltration, and a high rate of perineural invasion, contributing to significant morbidity and poor long-term outcomes. Given the absence of standardized treatment protocols, managing SEDC remains a challenge. Case Presentation: We report an unusual case of an 80-year-old female presenting with progressive numbness, nasal deviation, and a subcutaneous indurated lesion in the left nasofacial region. The early neurological symptoms were an atypical feature, suggesting perineural invasion (PNI) before visible tumor progression. Initial histopathologic evaluation was inconclusive, raising suspicion of SCC, necessitating immunohistochemical analysis, which confirmed ductal differentiation, leading to the final diagnosis of SEDC. The patient underwent radical resection with intraoperative margin assessment (Mohs micrographic surgery; MMS) followed by adjuvant radiotherapy (62 Gy/31 fractions) due to high-risk features, including perineural and perivascular invasion. Despite initial disease control, a local recurrence involving the left orbit and nasal bone occurred 20 months postoperatively, demonstrating the aggressive nature of SEDC despite clear surgical margins and adjuvant therapy. Due to disease progression and refusal of further surgery, only palliative care was provided. During follow-up, the patient contracted COVID-19, further complicating her clinical status and contributing to her demise. While COVID-19 was not directly linked to SEDC progression, its impact on patient management was significant. Conclusions: This case underscores the diagnostic and therapeutic challenges of SEDC, emphasizing the need for early suspicion, extensive histopathologic assessment, and aggressive multimodal treatment. The importance of multidisciplinary management-particularly in elderly and immunocompromised patients-and long-term surveillance due to high recurrence risk and PNI is crucial.
Collapse
Affiliation(s)
- Bruno Špiljak
- Department of Oral Medicine, University of Zagreb School of Dental Medicine, 10000 Zagreb, Croatia;
| | - Damir Sauerborn
- Department of Otorhinolaryngology and Head and Neck Surgery, General Hospital “Dr. Josip Benčević”, 35000 Slavonski Brod, Croatia; (D.S.); (B.G.B.)
| | - Matej Tomas
- Department of Dental Medicine, Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia;
| | - Brankica Gregorić Butina
- Department of Otorhinolaryngology and Head and Neck Surgery, General Hospital “Dr. Josip Benčević”, 35000 Slavonski Brod, Croatia; (D.S.); (B.G.B.)
| | - Ivana Mahovne
- Department of Pathology and Cytology, General Hospital “Dr. Josip Benčević”, 35000 Slavonski Brod, Croatia;
| | - Suzana Erić
- Department of Oncology, Clinical Hospital Centre, 31000 Osijek, Croatia;
| | - Bruno Vidaković
- Department of Dental Medicine, Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia;
| | - Stjepanka Lešić
- Department of Dental Medicine, Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia;
| |
Collapse
|
11
|
Gong S, Yang L, Xu M, Xiang M, Lang J, Zhang H, Shan Y. A combined gene signature model for predicting radiotherapy response and relapse-free survival in laryngeal squamous cell carcinoma. Cancer Cell Int 2025; 25:102. [PMID: 40102978 PMCID: PMC11916850 DOI: 10.1186/s12935-025-03739-5] [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: 06/27/2024] [Accepted: 03/08/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Radioresistance is a major challenge in radiotherapy for laryngeal squamous cell carcinoma (LSCC), and there is currently no effective method to predict radiosensitivity in LSCC patients. This study aimed to establish a prediction model for radiotherapy response based on gene expression. METHODS The datasets of LSCC were obtained from the ENT department of Shanghai Ruijin Hospital and The Cancer Genome Atlas (TCGA). Lasso regression and Cox regression were used to establish the prediction model based on gene expression. Weighted gene coexpression network analysis (WGCNA) was used to analyze the correlation between gene expression and clinical characteristics. RT-qPCR was used to detect gene expression in tumor tissue to verify the accuracy of the prediction model. RESULTS Using a cohort of LSCC cases receiving radiotherapy collected in the TCGA database, the 3 protein-coding genes (PCGs) signature model was identified for the first time as the predictor of relapse-free survival and radiosensitivity in LSCC patients. And we explored the potential clinical value of 3 PCGs and screened out 2 long non-coding RNAs (lncRNAs) potential associated with 3 PCGs. More importantly, the LSCC cases collected by our department were used to preliminarily verify the predictive power of the 3 PCGs signature model for the radiosensitivity of LSCC, and the significant correlation between the expression levels of the 3 PCGs and the 2 lncRNAs. CONCLUSION We successfully establish a radiosensitivity prediction model based on the 3 PCGs Riskscore, which provides a theoretical basis for the decision-making of LSCC treatment options. Meantime, we preliminarily screen the potential associated lncRNAs of the 3 PCGs for further basic and clinical research.
Collapse
Affiliation(s)
- Shiqi Gong
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Liyun Yang
- Department of Otolaryngology, Gongli Hospital of Pudong, Shanghai, 200135, China
| | - Meng Xu
- Department of Oncology, Changhai Hospital Affiliated to Navy Medical University, Shanghai, 200433, China
| | - Mingliang Xiang
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Juntian Lang
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hao Zhang
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Yamin Shan
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
12
|
Sant’Angelo D, Descamps G, Lecomte V, Stanicki D, Penninckx S, Dragan T, Van Gestel D, Laurent S, Journe F. Therapeutic Approaches with Iron Oxide Nanoparticles to Induce Ferroptosis and Overcome Radioresistance in Cancers. Pharmaceuticals (Basel) 2025; 18:325. [PMID: 40143107 PMCID: PMC11945075 DOI: 10.3390/ph18030325] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/15/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
The emergence of nanotechnology in medicine, particularly using iron oxide nanoparticles (IONPs), may impact cancer treatment strategies. IONPs exhibit unique properties, such as superparamagnetism, biocompatibility, and ease of surface modification, making them ideal candidates for imaging, and therapeutic interventions. Their application in targeted drug delivery, especially with traditional chemotherapeutic agents like cisplatin, has shown potential in overcoming limitations such as low bioavailability and systemic toxicity of chemotherapies. Moreover, IONPs, by releasing iron ions, can induce ferroptosis, a form of iron-dependent cell death, which offers a promising pathway to reverse radio- and chemoresistance in cancer therapy. In particular, IONPs demonstrate significant potential as radiosensitisers, enhancing the effects of radiotherapy by promoting reactive oxygen species (ROS) generation, lipid peroxidation, and modulating the tumour microenvironment to stimulate antitumour immune responses. This review explores the multifunctional roles of IONPs in radiosensitisation through ferroptosis induction, highlighting their promise in advancing treatment for head and neck cancers. Additional research is crucial to fully addressing their potential in clinical settings, offering a novel approach to personalised cancer treatment.
Collapse
Affiliation(s)
- Dorianne Sant’Angelo
- Department of Human Biology and Toxicology (Cancer Research Unit), Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium
- Laboratory of Clinical and Experimental Oncology (LOCE), Institute Jules Bordet, HUB, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Géraldine Descamps
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Valentin Lecomte
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Dimitri Stanicki
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Sébastien Penninckx
- Department of Medical Physics, Institut Jules Bordet, HUB, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium;
- Department of Radiotherapy, Institute Jules Bordet, Hopital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (T.D.); (D.V.G.)
| | - Tatiana Dragan
- Department of Radiotherapy, Institute Jules Bordet, Hopital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (T.D.); (D.V.G.)
| | - Dirk Van Gestel
- Department of Radiotherapy, Institute Jules Bordet, Hopital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (T.D.); (D.V.G.)
| | - Sophie Laurent
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Fabrice Journe
- Department of Human Biology and Toxicology (Cancer Research Unit), Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium
- Laboratory of Clinical and Experimental Oncology (LOCE), Institute Jules Bordet, HUB, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| |
Collapse
|
13
|
Wu J, Zhao X, Ren B, Duan X, Sun J. Radioresistance in Hepatocellular Carcinoma: Biological Bases and Therapeutic Implications. Int J Mol Sci 2025; 26:1839. [PMID: 40076465 PMCID: PMC11899467 DOI: 10.3390/ijms26051839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor with high morbidity and mortality. Radiotherapy technology is a common treatment modality that can be used in all stages of HCC. However, in some cases, radiotherapy fails in clinical practice mainly because of the patient's resistance to radiotherapy, creating a bottleneck for future breakthroughs. HCC radiosensitivity is primarily related to DNA double-strand break repair, cellular autophagy, cell cycle, cellular metabolism, and hypoxic environmental regulators. Therefore, a comprehensive understanding of its molecular mechanisms will be of immense importance in reversing HCC radioresistance. In this review, we provide a comprehensive overview of the mechanism of action of radiotherapy on HCC, the cellular and molecular basis of radiation resistance in HCC, related treatment modalities, and future prospects.
Collapse
Affiliation(s)
- Jianhui Wu
- Department of Radiation Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
- Medical School, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaofang Zhao
- Department of Radiation Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
- Medical School, Chinese PLA General Hospital, Beijing 100853, China
| | - Bowen Ren
- Medical School, Chinese PLA General Hospital, Beijing 100853, China
| | - Xuezhang Duan
- Department of Radiation Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
| | - Jing Sun
- Department of Radiation Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
- Medical School, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
14
|
Zheng S, Piao Y, Jung SN, Oh C, Lim MA, Nguyen Q, Shen S, Park SH, Cui S, Piao S, Kim YI, Kim JW, Won HR, Chang JW, Shan Y, Liu L, Koo BS. Gene Expression Alteration by Non-thermal Plasma-Activated Media Treatment in Radioresistant Head and Neck Squamous Cell Carcinoma. Clin Exp Otorhinolaryngol 2025; 18:73-87. [PMID: 39757757 PMCID: PMC11917201 DOI: 10.21053/ceo.2024.00238] [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: 08/14/2024] [Accepted: 01/05/2025] [Indexed: 01/07/2025] Open
Abstract
OBJECTIVES Head and neck squamous cell carcinoma (HNSCC) exhibits high recurrence rates, particularly in cases of radioresistant HNSCC (RR-HNSCC). Non-thermal plasma (NTP) therapy effectively suppresses the progression of HNSCC. However, the therapeutic mechanisms of NTP therapy in treating RR-HNSCC are not well understood. In this study, we explored the regulatory role of NTP in the RR-HNSCC signaling pathway and identified its signature genes. METHODS After constructing two RR-HNSCC cell lines, we prepared cell lysates from cells treated or not treated with NTP-activated media (NTPAM) and performed RNA sequencing to determine their mRNA expression profiles. Based on the RNA sequencing results, we identified differentially expressed genes (DEGs), followed by a bioinformatics analysis to identify candidate molecules potentially associated with NTPAM therapy for RR-HNSCC. RESULTS NTPAM reduced RR-HNSCC cell viability in vitro. RNA sequencing results indicated that NTPAM treatment activated the reactive oxygen species (ROS) pathway and induced ferroptosis in RR-HNSCC cell lines. Among the 1,924 genes correlated with radiation treatment, eight showed statistical significance in both the cell lines and The Cancer Genome Atlas (TCGA) cohort. Only five genes-ABCC3, DUSP16, PDGFB, RAF1, and THBS1-showed consistent results between the NTPAM data sequencing and TCGA data. LASSO regression analysis revealed that five genes were associated with cancer prognosis, with a hazard ratio of 2.26. In RR-HNSCC cells, NTPAM affected DUSP16, PDGFB, and THBS1 as activated markers within 6 hours, and this effect persisted for 12 hours. Furthermore, enrichment analysis indicated that these three DEGs were associated with the extracellular matrix, transforming growth factor-beta, phosphoinositide 3-kinase/protein kinase B, and mesenchymal-epithelial transition factor pathways. CONCLUSION NTPAM therapy exerts cytotoxic effects in RR-HNSCC cell lines by inducing specific ROS-mediated ferroptosis. DUSP16, PDGFB, and THBS1 were identified as crucial targets for reversing the radiation resistance induced by NTPAM therapy, providing insights into the mechanisms and clinical applications of NTPAM treatment in RR-HNSCC.
Collapse
Affiliation(s)
- Sicong Zheng
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Yudan Piao
- Dental Department, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - Seung-Nam Jung
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - Chan Oh
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Mi Ae Lim
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - QuocKhanh Nguyen
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Shan Shen
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Se-Hee Park
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Shengzhe Cui
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Shuyu Piao
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - Young Il Kim
- Department of Radiation Oncology, Chungnam National University Sejong Hospital, Sejong, Korea
| | - Ji Won Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Chungnam National University Sejong Hospital, Sejong, Korea
| | - Ho-Ryun Won
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Chungnam National University Sejong Hospital, Sejong, Korea
| | - Jae Won Chang
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - Yujuan Shan
- Department of Nutrition, Public Health and Management College, Wenzhou Medical University, Wenzhou, China
| | - Lihua Liu
- Department of Nutrition, Public Health and Management College, Wenzhou Medical University, Wenzhou, China
| | - Bon Seok Koo
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| |
Collapse
|
15
|
Giunco S, Del Mistro A, Morello M, Lidonnici J, Frayle H, Gori S, De Rossi A, Boscolo-Rizzo P. From infection to immortality: The role of HPV and telomerase in head and neck cancer. Oral Oncol 2025; 161:107169. [PMID: 39755000 DOI: 10.1016/j.oraloncology.2024.107169] [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] [Revised: 12/19/2024] [Accepted: 12/25/2024] [Indexed: 01/06/2025]
Abstract
Head and neck squamous cell carcinomas (HNSCCs) represent a heterogeneous group of malignancies with multifactorial aetiologies. High-risk human papillomavirus (hrHPV) infections, particularly HPV16, and the dysregulation of telomerase activity, specifically through its catalytic subunit, telomerase reverse transcriptase (TERT) are among the key contributors to HNSCC development and progression. HPV promotes oncogenesis via the E6 and E7 oncoproteins, which inactivate tumour suppressors TP53 and RB1, leading to unchecked cellular proliferation. Concurrently, telomerase activation plays a critical role in HNSCC by maintaining telomere length, thus enabling cellular immortality, and facilitating tumour development and progression. The interplay between HPV and telomerase is significant; HPV oncoprotein E6 enhances telomerase activity through multiple regulatory mechanisms, including upregulating TERT expression. Beyond telomere maintenance, TERT influences signalling pathways, cellular metabolism, and the tumour microenvironment, contributing to aggressive tumour behaviour and poor prognosis. This review integrates the roles of HPV and telomerase in HNSCC, focusing on their molecular mechanisms and interactions that drive carcinogenesis and influence disease progression. Understanding the synergistic effects of HPV and TERT in HNSCC may be crucial for risk stratification, prognostic assessment, and the development of novel therapeutic strategies targeting these specific molecular pathways.
Collapse
Affiliation(s)
- Silvia Giunco
- Department of Surgery, Oncology and Gastroenterology, Section of Oncology and Immunology, University of Padova, 35128 Padova, Italy; Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, 35128 Padova, Italy
| | - Annarosa Del Mistro
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, 35128 Padova, Italy
| | - Marzia Morello
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, 35128 Padova, Italy
| | - Jacopo Lidonnici
- Department of Surgery, Oncology and Gastroenterology, Section of Oncology and Immunology, University of Padova, 35128 Padova, Italy
| | - Helena Frayle
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, 35128 Padova, Italy
| | - Silvia Gori
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, 35128 Padova, Italy
| | - Anita De Rossi
- Department of Surgery, Oncology and Gastroenterology, Section of Oncology and Immunology, University of Padova, 35128 Padova, Italy.
| | - Paolo Boscolo-Rizzo
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy
| |
Collapse
|
16
|
Wang S, Jiang J, Xing M, Su H. Deciphering the prognostic potential of a necroptosis-related gene signature in head and neck squamous cell carcinoma: a bioinformatic analysis. Transl Cancer Res 2025; 14:340-353. [PMID: 39974401 PMCID: PMC11833364 DOI: 10.21037/tcr-24-743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 11/21/2024] [Indexed: 02/21/2025]
Abstract
Background Necroptosis, an alternative mode of programmed cell death (PCD) that overcomes apoptosis resistance, has been implicated in the progression and drug resistance of cancer. The aim of this study is to find the biological and prognostic significance of necroptosis in patients with head and neck squamous cell carcinoma (HNSCC). Methods Integrated clinical datasets from The Cancer Genome Atlas (TCGA) HNSCC cohort underwent analysis. R package "DESeq2" was used to conduct differential gene expression analysis between normal and tumor tissues in the cohort, resulting in the identification of 2,172 differentially expressed genes (DEGs). A total of 159 necroptosis-related genes (NRGs) were extracted and performed a Venn analysis to identify the optimal necroptosis-related DEGs, resulting in the selection of 25 genes specifically associated with necroptosis in HNSCC. Then prognostic analyze, Cox regression analysis and prognostic model were demonstrated the ability to predict the extent of immunological infiltration in HNSCC. Results Among these DEGs, five genes (FADD, H2AZ1, PYGL, JAK3, and ZBP1) were found to have prognostic value (P<0.05). Then, bioinformatic analyses were conducted, and the biological and clinical significance of these five genes were demonstrated. Furthermore, Cox regression analysis was performed to develop a prognostic gene model based on these genes, which effectively classified HNSCC patients into low- or high-risk groups. The prognostic model also demonstrated the ability to predict the extent of immunological infiltration in HNSCC. Additionally, a predictive nomogram based on the clinicopathological features of these five prognostic DEGs was constructed. Conclusions We performed a systematic bioinformatic analysis to identify necroptosis-related prognostic genes in HNSCC patients. These genes' prognostic value was synthesized into a predictive nomogram for forecasting HNSCC progression.
Collapse
Affiliation(s)
- Shizhe Wang
- Xuhui District Dental Center, Shanghai, China
| | | | - Min Xing
- Xuhui District Dental Center, Shanghai, China
| | - Hongru Su
- Xuhui District Dental Center, Shanghai, China
| |
Collapse
|
17
|
Li X, Escoffier H, Sauter T, Tavassoli M. Targeting Fibroblast-Derived Interleukin 6: A Strategy to Overcome Epithelial-Mesenchymal Transition and Radioresistance in Head and Neck Cancer. Cancers (Basel) 2025; 17:267. [PMID: 39858048 PMCID: PMC11763410 DOI: 10.3390/cancers17020267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/06/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Cancer-associated fibroblasts have been reported to play a central role in driving cancer progression, promoting metastasis, and conferring resistance to therapy in HNSCC. METHODS Indirect and direct co-culture models of HPV-positive and HPV-negative HNSCC cells with fibroblasts were developed to study the effect of fibroblasts on cancer cells. ELISA was used to measure IL-6 secretion in these models. To dissect the underlying signalling mechanisms, the effects of IL-6, an IL-6 receptor (IL-6R) inhibitor, a MAPK/ERK inhibitor, and a JAK/STAT inhibitor were evaluated. Epithelial-to-mesenchymal transition (EMT) was assessed by measuring EMT markers and conducting scratch assays and spheroid assays. Radioresistance was evaluated using clonogenic assays. Additionally, radioresistant (RR) cell lines were established from parental cells to examine the correlation between radioresistance and EMT. RESULTS Fibroblasts were found to drive EMT-like changes and heightened radioresistance in HNSCC cells through IL-6 secretion. Remarkably, these Fb-driven effects were robustly reversed using IL-6R and MAPK/ERK inhibitors in both HPV-positive and HPV-negative cell lines, whereas JAK/STAT inhibitors proved effective only in HPV-negative cells. RR cell lines exhibit a more aggressive phenotype than their parental counterparts, marked by pronounced EMT features and heightened resistance to radiotherapy. Importantly, these aggressive characteristics were substantially attenuated by targeting IL-6R or MAPK/ERK pathways. CONCLUSIONS This study highlights the critical role of fibroblast-secreted IL-6 in driving and maintaining EMT and radioresistance in HNSCC, resulting in a more aggressive tumour phenotype. Targeting the IL-6/IL-6R/ERK pathway emerges as a promising therapeutic approach for combating CAF-driven tumour progression and improving clinical outcomes in patients with aggressive, therapy-resistant HNSCC.
Collapse
Affiliation(s)
- Xinyang Li
- Head and Neck Oncology Group, Centre for Host Microbiome Interaction, King’s College London, Hodgkin Building, London SE1 1UL, UK;
| | - Hugues Escoffier
- Department of Life Sciences and Medicine, University of Luxembourg, L-4370 Belvaux, Luxembourg (T.S.)
| | - Thomas Sauter
- Department of Life Sciences and Medicine, University of Luxembourg, L-4370 Belvaux, Luxembourg (T.S.)
| | - Mahvash Tavassoli
- Head and Neck Oncology Group, Centre for Host Microbiome Interaction, King’s College London, Hodgkin Building, London SE1 1UL, UK;
| |
Collapse
|
18
|
Chan SPY, Yeo CPX, Hong BH, Tan EMC, Beh CY, Yeo ELL, Poon DJJ, Chu PL, Soo KC, Chua MLK, Chow EKH. Combinatorial functionomics identifies HDAC6-dependent molecular vulnerability of radioresistant head and neck cancer. Exp Hematol Oncol 2025; 14:5. [PMID: 39800760 PMCID: PMC11727331 DOI: 10.1186/s40164-024-00590-8] [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: 06/17/2024] [Accepted: 12/07/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Radiotherapy is the primary treatment modality for most head and neck cancers (HNCs). Despite the addition of chemotherapy to radiotherapy to enhance its tumoricidal effects, almost a third of HNC patients suffer from locoregional relapses. Salvage therapy options for such recurrences are limited and often suboptimal, partly owing to divergent tumor and microenvironmental factors underpinning radioresistance. In this study, we utilized a combinatorial functionomics approach, the Quadratic Phenotypic Optimization Platform (QPOP), to rationally design drug pairings that exploit the molecular fingerprint and vulnerability of established in vitro isogenic radioresistant (RR)-HNC models. METHODS A QPOP-specific protocol was applied to RR-HNC models to rank and compare all possible drug combinations from a 12-drug set comprising standard chemotherapy, small molecule inhibitors and targeted therapies specific to HNC. Drug combination efficacy was evaluated by computing combination index scores, and by measuring apoptotic response. Drug targeting was validated by western blot analyses, and the Comet assay was used to quantify DNA damage. Enhanced histone deacetylase inhibitor (HDACi) efficacy in RR models was further examined by in vivo studies, and genetic and chemical inhibition of major Class I/II HDACs. Regulatory roles of HDAC6/SP1 axis were investigated using immunoprecipitation, gel shift and ChIP-qPCR assays. Comparative transcriptomic analyses were employed to determine the prognostic significance of targeting HDAC6. RESULTS We report the therapeutic potential of combining panobinostat (pan-HDAC inhibitor) with AZD7762 (CHK1/2 inhibitor; AstraZeneca) or ionizing radiation (IR) to re-sensitize RR-HNC cells and showed increased DNA damage underlying enhanced synergy. We further refined this RR-specific drug combination and prioritized HDAC6 as a targetable dependency in reversing radioresistance. We provide mechanistic insights into HDAC6-mediated regulation via a crosstalk involving SP1 and oncogenic and repair genes. From two independent patient cohorts, we identified a four-gene signature that may have discriminative ability to predict for radioresistance and amenable to HDAC6 inhibition. CONCLUSION We have uncovered HDAC6 as a promising molecular vulnerability that should be explored to treat RR-HNC.
Collapse
Affiliation(s)
- Sharon Pei Yi Chan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Celestia Pei Xuan Yeo
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Boon Hao Hong
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Evelyn Mui Cheng Tan
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Chaw Yee Beh
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Eugenia Li Ling Yeo
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Dennis Jun Jie Poon
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Pek Lim Chu
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, Cancer and Stem Cell Biology Programme, Singapore, Singapore
| | - Khee Chee Soo
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Melvin Lee Kiang Chua
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore.
- Duke-NUS Medical School, Oncology Academic Programme, Singapore, Singapore.
- Department of Head and Neck and Thoracic Cancers, Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore.
| | - Edward Kai-Hua Chow
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
19
|
Barham WT, Stagg MP, Mualla R, DiLeo M, Kansara S. Recurrent and Metastatic Head and Neck Cancer: Mechanisms of Treatment Failure, Treatment Paradigms, and New Horizons. Cancers (Basel) 2025; 17:144. [PMID: 39796771 PMCID: PMC11720666 DOI: 10.3390/cancers17010144] [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: 11/27/2024] [Revised: 12/21/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Background: Head and neck cancer is a deadly disease with over 500,000 cases annually worldwide. Metastatic head and neck cancer accounts for a large proportion of the mortality associated with this disease. Many advances have been made in our understanding of the mechanisms of metastasis. The application of immunotherapy to locally recurrent or metastatic head and neck cancer has not only improved oncologic outcomes but has also provided valuable insights into the mechanisms of immune evasion and ultimately treatment failure. Objectives: This review paper will review our current understanding of biological mechanisms of treatment failure and metastasis. Published and ongoing clinical trials in the management of metastatic head and neck cancer will also be summarized. Methods: A narrative review was conducted to address the current understanding of the mechanisms of treatment failure and current treatment paradigms in recurrent and metastatic head and neck carcinoma. Conclusions: Our understanding of treatment failure in this disease is rapidly evolving. Immunotherapy represents a valuable new tool in the fight against recurrent and metastatic head and neck squamous cell carcinoma. Integrating patient and tumor specific data via artificial intelligence and deep learning will allow for a precision oncology approach, thereby achieving better prognostication and management of patients with this deadly disease.
Collapse
Affiliation(s)
- William T. Barham
- Department of Otolaryngology-Head and Neck Surgery, Louisiana State University Health Sciences Center, New Orleans, LA 71103, USA; (W.T.B.); (R.M.); (M.D.)
| | - Marshall Patrick Stagg
- Department of Oncology, Our Lady of the Lake Regional Medical Center, Baton Rouge, LA 70809, USA;
| | - Rula Mualla
- Department of Otolaryngology-Head and Neck Surgery, Louisiana State University Health Sciences Center, New Orleans, LA 71103, USA; (W.T.B.); (R.M.); (M.D.)
| | - Michael DiLeo
- Department of Otolaryngology-Head and Neck Surgery, Louisiana State University Health Sciences Center, New Orleans, LA 71103, USA; (W.T.B.); (R.M.); (M.D.)
| | - Sagar Kansara
- Department of Otolaryngology-Head and Neck Surgery, Louisiana State University Health Sciences Center, New Orleans, LA 71103, USA; (W.T.B.); (R.M.); (M.D.)
| |
Collapse
|
20
|
Liang L, Albers AE, Taube ET, Pohl J, Schmidt S, Greve M, Kaufmann AM. Identifying Truly HPV-Driven Head and Neck Squamous Cell Carcinoma by QuantiGene-Molecular-Profiling-Histology Assay Allows for More Precise Prognosis Prediction. Int J Mol Sci 2024; 25:13643. [PMID: 39769405 PMCID: PMC11728353 DOI: 10.3390/ijms252413643] [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: 11/01/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) with discordant diagnostic patterns of HPV+/p16- or HPV-/p16+ correlate with worse prognosis. This study aims to identify truly HPV-driven HNSCCs using a QuantiGene-Molecular-Profiling-Histology (QG-MPH) assay for identifying transcriptionally active HPV. Of 97 FFPE samples analyzed, 68 were valid for HPV DNA detection by PCR and quantification of HPV E7 and p16 INK4a mRNA by QG-MPH. p16 INK4a mRNA expression was compared with p16 protein expression via immunohistochemistry (p16 IHC). Among the 68 cases, 26 (38.2%) showed increased high-risk HPV E7 mRNA expression (hrHPV E7 mRNA+), while 37 (54.4%) were hrHPV DNA+. Concordance between HPV DNA and mRNA status was 70.1%. Notably, 79.2% of E7 mRNA+ cases were p16 IHC+, compared to 55.9% of DNA+ cases, demonstrating better concordance between HPV E7 mRNA+ status and p16 mRNA expression plus p16 IHC positivity. All patients (19/19) in the HPV E7 mRNA+/p16 IHC+ group survived the 5-year follow-up, compared to 59.5% (22/37) in the HPV E7 mRNA-/p16 IHC- group (p = 0.001). Specifically, the OS rate was 57.1% (8/14) in the group with discordant HPV DNA and p16 IHC results, compared to 40% (3/5) in the group with discordant HPV E7 mRNA and p16 IHC results. These findings highlight the better outcomes for the transcriptionally active HPV cases and indicate the prognostic disadvantage for patients with discordant patterns and the advantages for incorporating the molecular mRNA profiling by QG-MPH to p16 IHC. In conclusion, QG-MPH quantification of E7 and p16 INK4a mRNA more precisely identifies truly HPV-driven from non-HPV-driven HNSCC, compared to HPV DNA testing alone or with p16 IHC, which reduces misclassification and provides valuable implications for improved prognosis prediction and therapeutic decision-making.
Collapse
Affiliation(s)
- Lili Liang
- HPV Research Laboratory, Department for Gynecology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (L.L.)
| | - Andreas E. Albers
- HPV Research Laboratory, Department for Gynecology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (L.L.)
- Klinische Medizin, Schwerpunkt Hals-Nasen-Ohren-Heilkunde, MSB Medical School Berlin, Hochschule für Gesundheit und Medizin, Rüdesheimerstr. 50, 14197 Berlin, Germany
| | - Eliane T. Taube
- Institute for Pathology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jonathan Pohl
- Institute for Pathology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Stephanie Schmidt
- Klinische Medizin, Schwerpunkt Hals-Nasen-Ohren-Heilkunde, MSB Medical School Berlin, Hochschule für Gesundheit und Medizin, Rüdesheimerstr. 50, 14197 Berlin, Germany
| | - Marla Greve
- HPV Research Laboratory, Department for Gynecology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (L.L.)
| | - Andreas M. Kaufmann
- HPV Research Laboratory, Department for Gynecology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (L.L.)
| |
Collapse
|
21
|
Li Q, Zhang Y, Luo S, Zhang Z, Oberg AL, Kozono DE, Lu H, Sarkaria JN, Ma L, Wang L. Identify Non-mutational p53 Functional Deficiency in Human Cancers. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae064. [PMID: 39325855 PMCID: PMC11702981 DOI: 10.1093/gpbjnl/qzae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 07/23/2024] [Accepted: 08/21/2024] [Indexed: 09/28/2024]
Abstract
An accurate assessment of p53's functional statuses is critical for cancer genomic medicine. However, there is a significant challenge in identifying tumors with non-mutational p53 inactivation which is not detectable through DNA sequencing. These undetected cases are often misclassified as p53-normal, leading to inaccurate prognosis and downstream association analyses. To address this issue, we built the support vector machine (SVM) models to systematically reassess p53's functional statuses in TP53 wild-type (TP53WT) tumors from multiple The Cancer Genome Atlas (TCGA) cohorts. Cross-validation demonstrated the good performance of the SVM models with a mean area under the receiver operating characteristic curve (AUROC) of 0.9822, precision of 0.9747, and recall of 0.9784. Our study revealed that a significant proportion (87%-99%) of TP53WT tumors actually had compromised p53 function. Additional analyses uncovered that these genetically intact but functionally impaired (termed as predictively reduced function of p53 or TP53WT-pRF) tumors exhibited genomic and pathophysiologic features akin to TP53-mutant tumors: heightened genomic instability and elevated levels of hypoxia. Clinically, patients with TP53WT-pRF tumors experienced significantly shortened overall survival or progression-free survival compared to those with predictively normal function of p53 (TP53WT-pN) tumors, and these patients also displayed increased sensitivity to platinum-based chemotherapy and radiation therapy.
Collapse
Affiliation(s)
- Qianpeng Li
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Zhang
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sicheng Luo
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhang Zhang
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ann L Oberg
- Division of Computational Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - David E Kozono
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and Women’s Hospital, Boston, MA 02215, USA
| | - Hua Lu
- Department of Biochemistry & Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Lina Ma
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liguo Wang
- Division of Computational Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
- Bioinformatics and Computational Biology Graduate Program, University of Minnesota Rochester, Rochester, MN 55904, USA
| |
Collapse
|
22
|
Palazzo M, Novizio N, Belvedere R, Oliviero C, Pacelli R, Merolla F, Staibano S, Petrella A. Metformin radiosensitizes OSCC in 2D and 3D models: possible involvement of CAF-1. Pathologica 2024; 116:404-416. [PMID: 39748725 DOI: 10.32074/1591-951x-1085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
Objective This study investigated metformin as a sensitizer for radiotherapy in oral squamous cell carcinoma (OSCC) to reduce the radiation intensity. It evaluated the drug's effect on Chromatin Assembly Factor-1 (CAF-1) expression, whose high levels correlate with worse prognosis of this cancer. Methods The effects of metformin, alone and with radiotherapy, were evaluated on CAL27 (HPV-) and SCC154 (HPV+) OSCC cells. The analyses were performed on cell monolayers by colony-forming assay, motility, and confocal microscopy. In spheroid 3D models, the sensitizing effect of metformin was assessed by measuring areas. CAF-1 expression affected by metformin was evaluated via Western blot, and its role was investigated by siRNAs. Results Metformin reduced the cells' ability to form colonies, migrate and invade, and promoted the acquisition of a less aggressive phenotype by increased E-cadherin and decreased N-cadherin expressions. Moreover, metformin lowered the IC50 of radiotherapy and showed strong effects on spheroid growth. Metformin downmodulated the expression of the major subunits of CAF-1, and the knockdown of this protein by siRNAs elicited a metformin-like effect on cell aggressiveness. Conclusions Metformin emerged as a promising adjuvant drug in OSCC because of its effects on cell aggressiveness and radiosensitizing action. These activities could be CAF-1-mediated.
Collapse
Affiliation(s)
| | | | | | | | - Roberto Pacelli
- Department of Advanced Biomedical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Francesco Merolla
- Department of Medicine and Health Sciences V. Tiberio, University of Molise, Campobasso, Italy
| | - Stefania Staibano
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Italy
| | | |
Collapse
|
23
|
Lee HM, Kuo PC, Chen WH, Chen PJ, Lam SH, Su YC, Chen CH. Diterpenoid from Croton tonkinensis as a Potential Radiation Sensitizer in Oral Squamous Cell Carcinoma: An In Vitro Study. Int J Mol Sci 2024; 25:11839. [PMID: 39519390 PMCID: PMC11546982 DOI: 10.3390/ijms252111839] [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: 10/01/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Radiotherapy combined with a radiosensitizer represents an important treatment for head and neck squamous cell carcinoma (HNSCC). Only a few chemotherapy agents are currently approved as radiosensitizers for targeted therapy. Oral squamous cell carcinoma is one of the deadliest cancers, with approximately ~500,000 new diagnosed cases and 145,000 deaths worldwide per year. The incidence of new cases continues to increase in developing countries. This study aimed to investigate the effect of Croton tonkinensis and Curcuma longa on cell viability in OSCC cells. The HNSCC cell line OML1 and its radiation-resistant clone OML1-R were used. The anticancer effect and the mechanism of action of Croton tonkinensis and Curcuma longa in OSCC cells were analyzed by using cell viability assays, Western blot analysis, and Tranwell migration assays. The results showed that Croton tonkinensis concentration-dependently reduced the viability of OML1 and OML1-R (radioresistant) cells by downregulating the levels of AKT/mTOR mediators, such as p110α, p85, pAKT (ser473), p-mTOR (ser2448), and p-S6 Ribosomal (ser235/236). We found that cotreatment of OML1 and OML1R cells with either zVAD-FMK (apoptosis inhibitor), Ferrostatin-1 (Fer-1, a ferroptosis inhibitor), or chloroquine (CQ, an autophagy inhibitor) markedly reduced cell death. These results demonstrate that Croton tonkinensis exhibits anti-proliferation activity and highlight the therapeutic potential of small-molecule inhibitors against PI3K/mTOR signaling for radiosensitization in HNC treatment.
Collapse
Affiliation(s)
- Hui-Ming Lee
- Division of General Surgery, Department of Surgery, E-Da Cancer Hospital, I-Shou University, Kaohsiung 824005, Taiwan;
- School of Chinese Medicine for Post Baccalaureate, College of Medicine, I-Shou University, Kaohsiung 84001, Taiwan
| | - Ping-Chung Kuo
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; (P.-C.K.); (S.-H.L.)
| | - Wen-Hui Chen
- Department of Dentistry, E-Da Hospital, I-Shou University, Kaohsiung 824005, Taiwan;
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Po-Jen Chen
- Department of Medical Research, E-Da Hospital, Kaohsiung 824, Taiwan;
| | - Sio-Hong Lam
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; (P.-C.K.); (S.-H.L.)
| | - Yu-Chieh Su
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
- Division of Hematology-Oncology, Department of Internal Medicine, E-Da Hospital, Kaohsiung 824410, Taiwan
| | - Chih-Hao Chen
- Department of Thoracic Surgery, Mackay Memorial Hospital, Taipei 104, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City 252, Taiwan
| |
Collapse
|
24
|
Reinema FV, Hudson N, Adema GJ, Peeters WJM, Neuzil J, Stursa J, Werner L, Sweep FCGJ, Bussink J, Span PN. MitoTam induces ferroptosis and increases radiosensitivity in head and neck cancer cells. Radiother Oncol 2024; 200:110503. [PMID: 39186982 DOI: 10.1016/j.radonc.2024.110503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND AND PURPOSE Radiotherapy (RT) is an integral treatment part for patients with head and neck squamous cell carcinoma (HNSCC), but radioresistance remains a major issue. Here, we use MitoTam, a mitochondrially targeted analogue of tamoxifen, which we aim to stimulate ferroptotic cell death with, and sensitize radioresistant cells to RT. MATERIALS AND METHODS We assessed viability, reactive oxygen species (ROS) production, disruption of mitochondrial membrane potential, and lipid peroxidation in radiosensitive (UT-SCC-40) and radioresistant (UT-SCC-5) HNSCC cells following MitoTam treatment. To assess ferroptosis specificity, we used the ferroptosis inhibitor ferrostatin-1 (fer-1). Also, total antioxidant capacity and sensitivity to tert-butyl hydroperoxide were evaluated to assess ROS-responses. 53BP1 staining was used to assess radiosensitivity after MitoTam treatment. RESULTS Our data revealed increased ROS, cell death, disruption of mitochondrial membrane potential, and lipid peroxidation following MitoTam treatment in both cell lines. Adverse effects of MitoTam on cell death, membrane potential and lipid peroxidation were prevented by fer-1, indicating induction of ferroptosis. Radioresistant HNSCC cells were less sensitive to the effects of MitoTam due to intrinsic higher antioxidant capacity. MitoTam treatment prior to RT led to superadditive residual DNA damage expressed by 53BP1 foci compared to RT or MitoTam alone. CONCLUSION MitoTam induced ferroptosis in HNSCC cells, which could be used to overcome the elevated antioxidant capacity of radioresistant cells and sensitize such cells to RT. Treatment with MitoTam followed by RT could therefore present a promising effective therapy of radioresistant cancers. STATEMENT OF SIGNIFICANCE Radiotherapy is applied in the treatment of a majority of cancer patients. Radioresistance due to elevated antioxidant levels can be overcome by promoting ferroptotic cell death combining ROS-inducing drug MitoTam with radiotherapy.
Collapse
Affiliation(s)
- F V Reinema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen the Netherlands
| | - N Hudson
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen the Netherlands
| | - G J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen the Netherlands
| | - W J M Peeters
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen the Netherlands
| | - J Neuzil
- School of Pharmacy and Medical Science, Griffith University, Southport, QLD 4222, Australia; Faculty of Science and First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic; Institute of Biotechnology, Czech Academy of Sciences, Prague-West 252 50, Czech Republic
| | - J Stursa
- Faculty of Science and First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic; Institute of Biotechnology, Czech Academy of Sciences, Prague-West 252 50, Czech Republic
| | - L Werner
- Faculty of Science and First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic; Institute of Biotechnology, Czech Academy of Sciences, Prague-West 252 50, Czech Republic
| | - F C G J Sweep
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - J Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen the Netherlands
| | - P N Span
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen the Netherlands.
| |
Collapse
|
25
|
Rutihinda C, Haroun R, Ordonez JP, Mohssine S, Oweida H, Sharma M, Fares M, Ruiz-Dominguez N, Pacheco MFM, Naasri S, Saidi NE, Oweida AJ. Gingerol acts as a potent radiosensitizer in head and neck squamous cell carcinoma. Discov Oncol 2024; 15:553. [PMID: 39397185 PMCID: PMC11471747 DOI: 10.1007/s12672-024-01425-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/03/2024] [Indexed: 10/15/2024] Open
Abstract
Treatment options for advanced head and neck squamous cell carcinoma (HNSCC) are limited and often cause severe toxicity and debilitating long-term impacts. Developing effective and safer treatments is warranted. Several plant extracts have shown their effectiveness, but a comprehensive comparison between plant extracts in HNSCC has not been reported. Our aim was to investigate the effect of different plant extracts on the proliferation and viability of HNSCC cell lines. In addition, we investigated the efficacy of combining cytotoxic plant extracts with radiation. Since RT is a cornerstone in the treatment and management of HNSCC, it is desirable to enhance its efficacy through combination with cytotoxic agents that have minimal side effects. HNSCC cell lines were treated with various plant extracts at different concentrations. MTT assays were performed to identify the most potent anti-tumor plant extract. Colony-formation assays were performed to determine the radiosensitization effect. To investigate the effect on migration, transwell migration assays were performed. Annexin V staining was performed to analyze cell apoptosis. 6-gingerol resulted in the most significant dose-dependent inhibition in all cell lines compared to other plant extracts. Colony-formation assays showed a significant radiosensitizing effect when 6-gingerol was combined with radiation. In addition, the combination of 6-gingerol with radiation resulted in a significant decrease in HNSCC cell migration. Mechanistically, Annexin V staining showed that the combination of 6-gingerol and RT induces a synergistic apoptotic effect in MOC1, MOC2 and SCC9 cells compared to RT alone. In conclusion, 6-gingerol enhances the effect of radiation in HNSCC cell lines and could be a suitable candidate for combination therapy in HNSCC.
Collapse
Affiliation(s)
- Cleopatra Rutihinda
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Ryma Haroun
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Juan Pablo Ordonez
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Saad Mohssine
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Huda Oweida
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Muskaan Sharma
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Mohamed Fares
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Nancy Ruiz-Dominguez
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Maria Fernanda Meza Pacheco
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Sahar Naasri
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Nour Elhouda Saidi
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Ayman J Oweida
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada.
| |
Collapse
|
26
|
Lee MK, Woo SR, Noh JK, Min S, Kong M, Lee YC, Ko SG, Eun YG. Prognostic Significance of SASP-Related Gene Signature of Radiation Therapy in Head and Neck Squamous Cell Carcinoma. Mol Cancer Ther 2024; 23:1348-1359. [PMID: 38959066 DOI: 10.1158/1535-7163.mct-23-0738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/20/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024]
Abstract
In this study, we developed and validated the clinical significance of senescence-associated secretory phenotype (SASP)-related gene signature and explored its association with radiation therapy (RT) in patients with head and neck squamous cell carcinoma (HNSCC). First, we searched the three published review literature associated with SASP and selected all 81 genes to develop SASP-related gene signature. Then, 81 SASP-related genes were adapted to gene expression dataset from The Cancer Genome Atlas (TCGA). Patients with HNSCC of TCGA were classified into clusters 1 and 2 via unsupervised clustering according to SASP-related gene signature. Kaplan-Meier plot survival analysis showed that cluster 1 had a poorer prognosis than cluster 2 in 5-year overall survival and recurrence-free survival. Similarly, cluster 1 showed a worse prognosis than cluster 2 in three validation cohorts (E-MTAB-8588, FHCRC, and KHU). Cox proportional hazards regression observed that the SASP-related signature was an independent prognostic factor for patients with HNSCC. We also established a nomogram using a relevant clinical parameter and a risk score. Time-dependent receiver operating characteristic analysis was carried out to assess the accuracy of the prognostic risk model and nomogram. Senescence SASP-related gene signature was associated with the response to RT. Therefore, subsequent, in vitro experiments further validated the association between SASP-related gene signature and RT in HNSCC. In conclusion, we developed a SASP-related gene signature, which could predict survival of patients with HNSCC, and this gene signature provides new clinical evidence for the accurate diagnosis and targeted RT of HNSCC.
Collapse
Affiliation(s)
- Min Kyeong Lee
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Seon Rang Woo
- Department of Otolaryngology-Head and Neck Surgery, Kyung Hee University School of Medicine, Kyung Hee University Medical Center, Seoul, Korea
| | - Joo Kyung Noh
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Soonki Min
- Department of Radiation Oncology, Kyung Hee University School of Medicine, Kyung Hee University Medical Center, Seoul, Korea
| | - Moonkyoo Kong
- Department of Radiation Oncology, Kyung Hee University School of Medicine, Kyung Hee University Medical Center, Seoul, Korea
| | - Young Chan Lee
- Department of Otolaryngology-Head and Neck Surgery, Kyung Hee University School of Medicine, Kyung Hee University Medical Center, Seoul, Korea
| | - Seong-Gyu Ko
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Young-Gyu Eun
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Department of Otolaryngology-Head and Neck Surgery, Kyung Hee University School of Medicine, Kyung Hee University Medical Center, Seoul, Korea
| |
Collapse
|
27
|
Howard D, Turnbull T, Wilson P, Paterson DJ, Milanova V, Thierry B, Kempson I. Quantitative Single-Cell Comparison of Sensitization to Radiation and a Radiomimetic Drug for Diverse Gold Nanoparticle Coatings. SMALL SCIENCE 2024; 4:2400053. [PMID: 40212074 PMCID: PMC11935229 DOI: 10.1002/smsc.202400053] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/24/2024] [Indexed: 04/13/2025] Open
Abstract
Metal-based nanoparticles (NPs) have entered clinical use for enhancing radiotherapy, but the underlying mechanisms remain ambiguous. Herein, single-cell analysis of two cell lines in response to megavolt irradiation and a radiomimetic drug, neocarzinostatin (NCS) after coculture with gold NPs with different surface coatings, polyethylene glycol (AuPEG), PEG, and transferrin (AuT) or silica (AuSiO2), is reported. Different surface chemistry presents a major challenge for objective comparison between the biological impacts where major differences in cell-uptake exist. AuSiO2 NPs are the most efficient for promoting radiosensitization despite being associated with cells 10 times less than the actively targeted AuT NPs. Conversely, for cells exposed to NCS, AuSiO2 NPs impede the radiomimetic action and promote cell survival. AuT NPs enhance death of cells in combination with NCS showing that NPs can sensitize against cytotoxic agents in addition to radiation. While NPs contribute to radiosensitization (or enhancing/impeding chemotherapeutic drug activity), due to cell and cell line heterogeneity, the ultimate radiosensitivity of a cell appears to be dominated by its inherent radiosensitivity and how this cell-regulated response is manipulated by NPs. This is evidenced through comparison of radiobiological response of cells with equivalent NP association rather than equivalent coculture conditions.
Collapse
Affiliation(s)
- Douglas Howard
- Future Industries InstituteUniversity of South AustraliaMawson LakesSouth Australia5095Australia
- Department of Nuclear MedicineUniversity Hospital EssenHufelandstrasse 5545122EssenGermany
| | - Tyron Turnbull
- Future Industries InstituteUniversity of South AustraliaMawson LakesSouth Australia5095Australia
| | - Puthenparampil Wilson
- UniSA STEMUniversity of South AustraliaMawson LakesSouth Australia5095Australia
- Department of Radiation OncologyRoyal Adelaide HospitalAdelaideSouth Australia5000Australia
| | | | - Valentina Milanova
- Future Industries InstituteUniversity of South AustraliaMawson LakesSouth Australia5095Australia
| | - Benjamin Thierry
- Future Industries InstituteUniversity of South AustraliaMawson LakesSouth Australia5095Australia
| | - Ivan Kempson
- Future Industries InstituteUniversity of South AustraliaMawson LakesSouth Australia5095Australia
| |
Collapse
|
28
|
Jassi C, kuo WW, Kuo CH, Chang CM, Chen MC, Shih TC, Li CC, Huang CY. Mediation of radiation-induced bystander effect and epigenetic modification: The role of exosomes in cancer radioresistance. Heliyon 2024; 10:e34460. [PMID: 39114003 PMCID: PMC11304029 DOI: 10.1016/j.heliyon.2024.e34460] [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: 12/21/2023] [Revised: 03/20/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
Exosomes are nano-sized extracellular vesicles produced by almost all mammalian cells. They play an important role in cell-to-cell communication by transferring biologically active molecules from the cell of origin to the recipient cells. Ionizing radiation influences exosome production and molecular cargo loading. In cancer management, ionizing radiation is a form of treatment that exerts its cancer cytotoxicity by induction of DNA damage and other alterations to the targeted tissue cells. However, normal bystander non-targeted cells may exhibit the effects of ionizing radiation, a phenomenon called radiation-induced bystander effect (RIBE). The mutual communication between the two groups of cells (targeted and non-targeted) via radiation-influenced exosomes enables the exchange of radiosensitive molecules. This facilitates indirect radiation exposure, leading, among other effects, to epigenetic remodeling and subsequent adaptation to radiation. This review discusses the role exosomes play in epigenetically induced radiotherapy resistance through the mediation of RIBE.
Collapse
Affiliation(s)
- Chikondi Jassi
- Department of Biological Sciences and Technology, China Medical University, Taichung, Taiwan
| | - Wei-Wen kuo
- Department of Biological Sciences and Technology, China Medical University, Taichung, Taiwan
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
- Department of Kinesiology and Health Science, College of William and Mary, Williamsburg, VA, USA
| | - Chun-Ming Chang
- Department of General Surgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Medicine Tzu Chi University, 701, Section 3, Chung-Yang Road, Hualien 97004, Taiwan
| | - Ming-Cheng Chen
- Division of Colorectal Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung 40705, Taiwan
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Tzu-Ching Shih
- Department of Biomedical Imaging & Radiological Science College of Medicine, China Medical University, Taichung 404, Taiwan
| | - Chi-Cheng Li
- School of Medicine Tzu Chi University, 701, Section 3, Chung-Yang Road, Hualien 97004, Taiwan
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Center of Stem Cell & Precision Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondria Related Diseases Research Center, Hualien Tzu Chi Hospital, Hualien 970, Taiwan
- Graduate Institute of Biomedicine, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung 413, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien 970, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
29
|
Mani L, Naveed A, McAdoo A, Rosenthal E, Hom M. Efficacy of depatuxizumab mafodotin (ABT-414) in preclinical models of head and neck cancer. Carcinogenesis 2024; 45:520-526. [PMID: 38375733 DOI: 10.1093/carcin/bgae014] [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: 11/05/2023] [Revised: 02/04/2024] [Accepted: 02/16/2024] [Indexed: 02/21/2024] Open
Abstract
Epidermal growth factor receptor (EGFR) is highly expressed in 80-90% of head and neck squamous cell carcinomas (HNSCCs), making it an ideal target for antibody-drug conjugates. Depatuxizumab mafodotin (ABT-414), is an EGFR-targeting ADC comprised of the monoclonal antibody ABT-806 conjugated to monomethyl auristatin F, a tubulin polymerization inhibitor. This study assessed the in vivo efficacy of ABT-414 in HNSCC. The effects of ABT-414 on HNSCCs were determined using in vitro cytotoxicity assays and in vivo flank xenograft mouse models. The distribution of ABT-414 was assessed ex vivo via optical imaging methods using a conjugate of ABT-414 to the near-infrared agent IRDye800. In vitro treatment of high EGFR-expressing human HNSCC cell lines (UMSCC47 and FaDu) with ABT-414 (0-3.38 nM) resulted in dose-dependent cell death (IC50 values of 0.213 nM and 0.167 nM, respectively). ABT-414 treatment of the FaDu mouse xenografts displayed antitumor activity (P = 0.023) without a change in body mass (P = 0.1335), whereas treatment of UMSCC47 did not generate a significant response (P = 0.1761). Fluorescence imaging revealed ABT-414-IRDye800 accumulation in the tumors of both FaDu and UMSCC47 cell lines, with a signal-to-background ratio of >10. ABT-414 treatment yielded antitumor activity in FaDu tumors, but not in UMSCC47, highlighting the potential for ABT-414 efficacy in high EGFR-expressing tumors. Although ABT-414-IRDye800 localized tumors in both cell lines, the differing antitumor responses highlight the need for further investigation into the role of the tumor microenvironment in drug delivery.
Collapse
Affiliation(s)
- Lucas Mani
- Department of Otolaryngology, Vanderbilt University, Nashville, USA
| | - Abdullah Naveed
- Department of Otolaryngology, Vanderbilt University, Nashville, USA
| | - Ashtyn McAdoo
- Department of Otolaryngology, Vanderbilt University, Nashville, USA
| | - Eben Rosenthal
- Department of Otolaryngology, Vanderbilt University, Nashville, USA
| | - Marisa Hom
- Department of Otolaryngology, Vanderbilt University, Nashville, USA
| |
Collapse
|
30
|
Yu M, Ni M, Xu F, Liu C, Chen L, Li J, Xia S, Diao Y, Chen J, Zhu J, Wu X, Tang M, Li J, Ke G. NSUN6-mediated 5-methylcytosine modification of NDRG1 mRNA promotes radioresistance in cervical cancer. Mol Cancer 2024; 23:139. [PMID: 38970106 PMCID: PMC11225205 DOI: 10.1186/s12943-024-02055-2] [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: 05/05/2024] [Accepted: 06/29/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Radioresistance is the leading cause of death in advanced cervical cancer (CC). Dysregulation of RNA modification has recently emerged as a regulatory mechanism in radiation and drug resistance. We aimed to explore the biological function and clinical significance of 5-methylcytosine (m5C) in cervical cancer radiosensitivity. METHODS The abundance of RNA modification in radiotherapy-resistant and sensitive CC specimens was quantified by liquid chromatography-tandem mass spectrometry. The essential RNA modification-related genes involved in CC radiosensitivity were screened via RNA sequencing. The effect of NSUN6 on radiosensitivity was verified in CC cell lines, cell-derived xenograft (CDX), and 3D bioprinted patient-derived organoid (PDO). The mechanisms of NSUN6 in regulating CC radiosensitivity were investigated by integrative m5C sequencing, mRNA sequencing, and RNA immunoprecipitation. RESULTS We found a higher abundance of m5C modification in resistant CC samples, and NSUN6 was the essential m5C-regulating gene concerning radiosensitivity. NSUN6 overexpression was clinically correlated with radioresistance and poor prognosis in cervical cancer. Functionally, higher NSUN6 expression was associated with radioresistance in the 3D PDO model of cervical cancer. Moreover, silencing NSUN6 increased CC radiosensitivity in vivo and in vitro. Mechanistically, NDRG1 was one of the downstream target genes of NSUN6 identified by integrated m5C-seq, mRNA-seq, and functional validation. NSUN6 promoted the m5C modification of NDRG1 mRNA, and the m5C reader ALYREF bound explicitly to the m5C-labeled NDRG1 mRNA and enhanced NDRG1 mRNA stability. NDRG1 overexpression promoted homologous recombination-mediated DNA repair, which in turn led to radioresistance in cervical cancer. CONCLUSIONS Aberrant m5C hypermethylation and NSUN6 overexpression drive resistance to radiotherapy in cervical cancer. Elevated NSUN6 expression promotes radioresistance in cervical cancer by activating the NSUN6/ALYREF-m5C-NDRG1 pathway. The low expression of NSUN6 in cervical cancer indicates sensitivity to radiotherapy and a better prognosis.
Collapse
Affiliation(s)
- Min Yu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Mengdong Ni
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Fei Xu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Chaohua Liu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Lihua Chen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jiana Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Siyu Xia
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yixin Diao
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jiaxin Chen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jun Zhu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Xiaohua Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Min Tang
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Jiajia Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Guihao Ke
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| |
Collapse
|
31
|
Pachler KS, Lauwers I, Verkaik NS, Rovituso M, van der Wal E, Mast H, Jonker BP, Sewnaik A, Hardillo JA, Keereweer S, Monserez D, Kremer B, Koppes S, van den Bosch TPP, Verduijn GM, Petit S, Sørensen BS, van Gent DC, Capala ME. Development of an Ex Vivo Functional Assay for Prediction of Irradiation Related Toxicity in Healthy Oral Mucosa Tissue. Int J Mol Sci 2024; 25:7157. [PMID: 39000262 PMCID: PMC11241643 DOI: 10.3390/ijms25137157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Radiotherapy in the head-and-neck area is one of the main curative treatment options. However, this comes at the cost of varying levels of normal tissue toxicity, affecting up to 80% of patients. Mucositis can cause pain, weight loss and treatment delays, leading to worse outcomes and a decreased quality of life. Therefore, there is an urgent need for an approach to predicting normal mucosal responses in patients prior to treatment. We here describe an assay to detect irradiation responses in healthy oral mucosa tissue. Mucosa specimens from the oral cavity were obtained after surgical resection, cut into thin slices, irradiated and cultured for three days. Seven samples were irradiated with X-ray, and three additional samples were irradiated with both X-ray and protons. Healthy oral mucosa tissue slices maintained normal morphology and viability for three days. We measured a dose-dependent response to X-ray irradiation and compared X-ray and proton irradiation in the same mucosa sample using standardized automated image analysis. Furthermore, increased levels of inflammation-inducing factors-major drivers of mucositis development-could be detected after irradiation. This model can be utilized for investigating mechanistic aspects of mucositis development and can be developed into an assay to predict radiation-induced toxicity in normal mucosa.
Collapse
Affiliation(s)
- Katrin S. Pachler
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (K.S.P.); (D.C.v.G.)
| | - Iris Lauwers
- Department of Radiotherapy, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Nicole S. Verkaik
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (K.S.P.); (D.C.v.G.)
| | - Marta Rovituso
- Holland Proton Therapy Centre (HPTC), Huismansingel 4, 2629 JH Delft, The Netherlands
| | - Ernst van der Wal
- Holland Proton Therapy Centre (HPTC), Huismansingel 4, 2629 JH Delft, The Netherlands
| | - Hetty Mast
- Department of Oral and Maxillofacial Surgery, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Brend P. Jonker
- Department of Oral and Maxillofacial Surgery, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Aniel Sewnaik
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Jose A. Hardillo
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Stijn Keereweer
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Dominiek Monserez
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Bernd Kremer
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Sjors Koppes
- Department of Pathology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | | | - Gerda M. Verduijn
- Department of Radiotherapy, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Steven Petit
- Department of Radiotherapy, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Brita S. Sørensen
- Department of Radiotherapy, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
- Department of Experimental Clinical Oncology, Danish Centre for Particle Therapy, Aarhus University Hospital, 8200 Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Nordre Ringgade 1, 8000 Aarhus, Denmark
| | - Dik C. van Gent
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (K.S.P.); (D.C.v.G.)
| | - Marta E. Capala
- Department of Radiotherapy, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
32
|
Gauss C, Stone LD, Ghafouri M, Quan D, Johnson J, Fribley AM, Amm HM. Overcoming Resistance to Standard-of-Care Therapies for Head and Neck Squamous Cell Carcinomas. Cells 2024; 13:1018. [PMID: 38920648 PMCID: PMC11201455 DOI: 10.3390/cells13121018] [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: 05/29/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
Although there have been some advances during in recent decades, the treatment of head and neck squamous cell carcinoma (HNSCC) remains challenging. Resistance is a major issue for various treatments that are used, including both the conventional standards of care (radiotherapy and platinum-based chemotherapy) and the newer EGFR and checkpoint inhibitors. In fact, all the non-surgical treatments currently used for HNSCC are associated with intrinsic and/or acquired resistance. Herein, we explore the cellular mechanisms of resistance reported in HNSCC, including those related to epigenetic factors, DNA repair defects, and several signaling pathways. This article discusses these mechanisms and possible approaches that can be used to target different pathways to sensitize HNSCC to the existing treatments, obtain better responses to new agents, and ultimately improve the patient outcomes.
Collapse
Affiliation(s)
- Chester Gauss
- Carman and Ann Adams Department of Pediatrics, School of Medicine, Wayne State University, Detroit, MI 48202, USA; (C.G.); (M.G.)
| | - Logan D. Stone
- Oral & Maxillofacial Surgery, School of Dentistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Mehrnoosh Ghafouri
- Carman and Ann Adams Department of Pediatrics, School of Medicine, Wayne State University, Detroit, MI 48202, USA; (C.G.); (M.G.)
| | - Daniel Quan
- Department of Otolaryngology Head and Neck Surgery, School of Medicine, Wayne State University, Detroit, MI 48202, USA; (D.Q.)
| | - Jared Johnson
- Department of Otolaryngology Head and Neck Surgery, School of Medicine, Wayne State University, Detroit, MI 48202, USA; (D.Q.)
| | - Andrew M. Fribley
- Carman and Ann Adams Department of Pediatrics, School of Medicine, Wayne State University, Detroit, MI 48202, USA; (C.G.); (M.G.)
- Department of Otolaryngology Head and Neck Surgery, School of Medicine, Wayne State University, Detroit, MI 48202, USA; (D.Q.)
- Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48202, USA
| | - Hope M. Amm
- Oral & Maxillofacial Surgery, School of Dentistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| |
Collapse
|
33
|
Geyer F, Geyer M, Reuning U, Klapproth S, Wolff KD, Nieberler M. CHD4 acts as a prognostic factor and drives radioresistance in HPV negative HNSCC. Sci Rep 2024; 14:8286. [PMID: 38594331 PMCID: PMC11003975 DOI: 10.1038/s41598-024-58958-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: 11/27/2023] [Accepted: 04/04/2024] [Indexed: 04/11/2024] Open
Abstract
Despite great efforts in improving existing therapies, the outcome of patients with advanced radioresistant HPV-negative head and neck squamous cell carcinoma (HNSCC) remains poor. The chromatin remodeler Chromodomain helicase DNA binding protein 4 (CHD4) is involved in different DNA-repair mechanisms, but the role and potential in HNSCC has not been explored yet. In the present study, we evaluated the prognostic significance of CHD4 expression using in silico analysis of the pan-cancer dataset. Furthermore, we established a monoclonal HNSCC CHD4 knockdown cell clone utilizing the CRISPR/Cas9 system. Effects of lower CHD4 expression on radiosensitivity after increasing doses of ionizing radiation were characterized using clonogenic assays and cell numbers. The in silico analysis revealed that high CHD4 expression is associated with significant poorer overall survival of HPV-negative HNSCC patients. Additionally, the knockdown of CHD4 significantly increased the radiosensitivity of HNSCC cells. Therefore, CHD4 might be involved in promoting radioresistance in hard-to-treat HPV-negative HNSCC entities. We conclude that CHD4 could serve as a prognostic factor in HPV-negative HNSCC tumors and is a potential target protein overcoming radioresistance in HNSCC. Our results and the newly established cell clone laid the foundation to further characterize the underlying mechanisms and ultimately use CHD4 in HNSCC therapies.
Collapse
Affiliation(s)
- Fabian Geyer
- Department of Oral and Maxillofacial Surgery, Klinikum Rechts der Isar der Technischen Universität München, 81675, Munich, Germany.
| | - Maximilian Geyer
- Department of Oral and Maxillofacial Surgery, Klinikum Rechts der Isar der Technischen Universität München, 81675, Munich, Germany
| | - Ute Reuning
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675, Munich, Germany
| | - Sarah Klapproth
- Institute of Experimental Hematology, School of Medicine, Technische Universität München, 81675, Munich, Germany
| | - Klaus-Dietrich Wolff
- Department of Oral and Maxillofacial Surgery, Klinikum Rechts der Isar der Technischen Universität München, 81675, Munich, Germany
| | - Markus Nieberler
- Department of Oral and Maxillofacial Surgery, Klinikum Rechts der Isar der Technischen Universität München, 81675, Munich, Germany
| |
Collapse
|
34
|
Ivers JD, Puvvada N, Quick CM, Rajaram N. Investigating the relationship between hypoxia, hypoxia-inducible factor 1, and the optical redox ratio in response to radiation therapy. BIOPHOTONICS DISCOVERY 2024; 1:015003. [PMID: 40109884 PMCID: PMC11922545 DOI: 10.1117/1.bios.1.1.015003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Significance Radiation resistance is a major contributor to cancer treatment failure and is likely driven by multiple pathways. Multivariate visualization that preserves the spatial co-localization of factors could aid in understanding mechanisms of resistance and identifying biomarkers of response. Aim We aim to investigate the spatial and temporal relationship between hypoxia, hypoxia-inducible factor 1 (HIF-1α), and metabolism in response to radiation therapy in two cell lines of known radiation resistance and sensitivity. Approach Two-photon excited fluorescence and fluorescence lifetime imaging microscopy were used to quantify the optical redox ratio (ORR) and NAD(P)H fluorescent lifetime and bound fraction in frozen tumor sections and co-registered with immunohistochemical stain-based imaging of hypoxic fraction and HIF-1α. Results Histogram analysis of hypoxia, HIF-1α, and ORR revealed an increase in the ORR in regions of low hypoxia and high HIF-1α, indicating that the stabilization of HIF-1α is likely due to an increase in reactive oxygen species following radiation therapy. In addition, the bound NAD(P)H fraction was higher in regions with a low ORR in resistant tumors following radiation, suggesting an increase in fatty acid synthesis. Conclusions A multivariate histogram approach can reveal hidden trends not observed in bulk analysis of tumor images and may be useful in understanding biomarkers and mechanisms of radiation resistance.
Collapse
Affiliation(s)
- Jesse D Ivers
- University of Arkansas, Department of Biomedical Engineering, Fayetteville, Arkansas, United States
| | - Nagavenkatasai Puvvada
- University of Arkansas, Department of Biomedical Engineering, Fayetteville, Arkansas, United States
| | - Charles M Quick
- University of Arkansas for Medical Sciences, Department of Pathology, Little Rock, Arkansas, United States
| | - Narasimhan Rajaram
- University of Arkansas, Department of Biomedical Engineering, Fayetteville, Arkansas, United States
| |
Collapse
|
35
|
Ma J, Na S, Wang P, Li J, He S, Liu F. miR-626 Inhibition Enhanced the Radiosensitivity to Oral Squamous Cell Carcinoma via the Downregulation of Nuclear Factor Kappa-B Signaling. Cancer Biother Radiopharm 2024; 39:144-152. [PMID: 35549438 DOI: 10.1089/cbr.2021.0344] [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: 11/13/2022] Open
Abstract
Objective: The effect of miR-626 on the radiosensitivity to oral squamous cell carcinoma (OSCC) was evaluated in this study. Materials and Methods: The level of miR-626 in OSCC patients was determined by analyzing the data of miRNA microarray GSE113956. miR-626 was overexpressed by miR-626 mimics and knockdown were performed by miR-626 inhibitor. The level of miR-626 was detected by quantitative real-time polymerase chain reaction. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and colony formation assays were used to detect the effect of miR-626 on the growth of OSCC cells. Flow cytometry was used to detect the apoptosis of OSCC cells. Western blot and dual luciferase reporter assays were used to explore the underlying mechanism of miR-626 regulating the radiosensitivity to OSCC. The effect of miR-626 on the radiosensitivity to OSCC were examined in an in vivo xenograft model. Results: The serum miR-626 level of OSCC patients was significantly higher than that of healthy controls. miR-626 mimics significantly promoted the OSCC cell growth, but the miR-626 inhibitor significantly suppressed the OSCC cell growth. Radiation combined with the miR-626 inhibitor significantly suppressed the cell proliferation and promoted the apoptosis of SCC-4 and HSC4 cells. Moreover, miR-626 regulates the nuclear factor kappa-B (NF-κB) signaling mediated by TRAF-interacting protein with forkhead-associated domain B. Furthermore, inhibition of miR-626 enhances the radiosensitivity to OSCC in nude mice. Conclusions: miR-626 inhibition enhanced the radiosensitivity to OSCC through the downregulation of NF-κB signaling.
Collapse
Affiliation(s)
- Jing Ma
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medical Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, P.R. China
- Department of Endodontics and Affiliated Stomatology Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Sijia Na
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medical Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, P.R. China
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Panxi Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medical Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Jinyi Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medical Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Shuyang He
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong
| | - Fei Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medical Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, P.R. China
- Department of Pediatric Dentistry, Affiliated Stomatology Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
36
|
Naumann F, Kaanders J, Peeters W, Adema G, Sweep F, Bussink J, Span P. Radiotherapy induces an increase in serum antioxidant capacity reflecting tumor response. Clin Transl Radiat Oncol 2024; 45:100726. [PMID: 38292333 PMCID: PMC10825560 DOI: 10.1016/j.ctro.2024.100726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/22/2023] [Accepted: 01/10/2024] [Indexed: 02/01/2024] Open
Abstract
Background and purpose Radiotherapy (RT) is a mainstay component of treatment for patients with head and neck squamous cell carcinoma (HNSCC), but responses vary. As RT relies upon oxidative damage, antioxidant expression in response to RT-induced reactive oxygen species (ROS) could compromise treatment response. We aimed to examine local and systemic antioxidant responses to increased RT-induced ROS in relation to treatment success. Materials and methods Nuclear factor erythroid 2-related factor 2 (NRF2), the main antioxidant transcription factor, was immunofluorescently stained in FaDu cells and in tumor biopsies of patients with oral cavity/oropharynx HNSCC before and after five fractions of RT. Besides, total antioxidant capacity (TAC) was analyzed in HNSCC tumor cells in vitro and in serum of HNSCC patients before, during, and after RT. Results Data revealed an increase in NRF2 expression and TAC in head and neck cancer cells in vitro over the course of 5 daily fractions of 2 Gy. In accordance, also in patients' tumors NRF2 expression increased, which was associated with increased serum TAC during RT. Increasing serum TAC was related to impaired local tumor control. Conclusion Radiation induced NRF2 expression and upregulated TAC, which may compromise the effect of RT-induced ROS. Changes in serum TAC during RT could serve as a novel predictor of treatment outcome in HNSCC patients.Medical Ethics Review Committee (CMO) approval - CMO number: 2007/104.
Collapse
Affiliation(s)
- F.V. Naumann
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - J.H.A.M. Kaanders
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - W.J.M. Peeters
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - G.J. Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - F.C.G.J. Sweep
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - J. Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - P.N. Span
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
37
|
Peña-Oyarzún D, Flores T, Torres VA, Quest AFG, Lobos-González L, Kretschmar C, Contreras P, Maturana-Ramírez A, Criollo A, Reyes M. Inhibition of PORCN Blocks Wnt Signaling to Attenuate Progression of Oral Carcinogenesis. Clin Cancer Res 2024; 30:209-223. [PMID: 37812478 DOI: 10.1158/1078-0432.ccr-23-0318] [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: 02/01/2023] [Revised: 05/12/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023]
Abstract
PURPOSE Oral squamous cell carcinoma (OSCC) is commonly preceded by potentially malignant lesions, referred to as oral dysplasia. We recently reported that oral dysplasia is associated with aberrant activation of the Wnt/β-catenin pathway, due to overexpression of Wnt ligands in a Porcupine (PORCN)-dependent manner. Pharmacologic inhibition of PORCN precludes Wnt secretion and has been proposed as a potential therapeutic approach to treat established cancers. Nevertheless, there are no studies that explore the effects of PORCN inhibition at the different stages of oral carcinogenesis. EXPERIMENTAL DESIGN We performed a model of tobacco-induced oral cancer in vitro, where dysplastic oral keratinocytes (DOK) were transformed into oral carcinoma cells (DOK-TC), and assessed the effects of inhibiting PORCN with the C59 inhibitor. Similarly, an in vivo model of oral carcinogenesis and ex vivo samples derived from patients diagnosed with oral dysplasia and OSCC were treated with C59. RESULTS Both in vitro and ex vivo oral carcinogenesis approaches revealed decreased levels of nuclear β-catenin and Wnt3a, as observed by immunofluorescence and IHC analyses. Consistently, reduced protein and mRNA levels of survivin were observed after treatment with C59. Functionally, treatment with C59 in vitro resulted in diminished cell migration, viability, and invasion. Finally, by using an in vivo model of oral carcinogenesis, we found that treatment with C59 prevented the development of OSCC by reducing the size and number of oral tumor lesions. CONCLUSIONS The inhibition of Wnt ligand secretion with C59 represents a feasible treatment to prevent the progression of early oral lesions toward OSCC.
Collapse
Affiliation(s)
- Daniel Peña-Oyarzún
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Physiology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Interdisciplinary Center for Research in Territorial Health of the Aconcagua Valley (CIISTe Aconcagua), School of Medicine, Faculty of Medicine, San Felipe Campus, Universidad de Valparaiso, Chile
| | - Tania Flores
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Research Centre in Dental Science (CICO), Faculty of Dentistry, Universidad de La Frontera, Temuco, Chile
- Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Vicente A Torres
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Andrew F G Quest
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Cellular Communication, Center for studies on Exercise, Metabolism and Cancer (CEMC), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Lorena Lobos-González
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Catalina Kretschmar
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Pamela Contreras
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Cellular Communication, Center for studies on Exercise, Metabolism and Cancer (CEMC), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Andrea Maturana-Ramírez
- Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Alfredo Criollo
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Montserrat Reyes
- Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| |
Collapse
|
38
|
Viswanath D, Park J, Misra R, Pizzuti VJ, Shin SH, Doh J, Won YY. Nanotechnology-enhanced radiotherapy and the abscopal effect: Current status and challenges of nanomaterial-based radio-immunotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1924. [PMID: 37632203 DOI: 10.1002/wnan.1924] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/03/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023]
Abstract
Rare but consistent reports of abscopal remission in patients challenge the notion that radiotherapy (RT) is a local treatment; radiation-induced cancer cell death can trigger activation and recruitment of dendritic cells to the primary tumor site, which subsequently initiates systemic immune responses against metastatic lesions. Although this abscopal effect was initially considered an anomaly, combining RT with immune checkpoint inhibitor therapies has been shown to greatly improve the incidence of abscopal responses via modulation of the immunosuppressive tumor microenvironment. Preclinical studies have demonstrated that nanomaterials can further improve the reliability and potency of the abscopal effect for various different types of cancer by (1) altering the cell death process to be more immunogenic, (2) facilitating the capture and transfer of tumor antigens from the site of cancer cell death to antigen-presenting cells, and (3) co-delivering immune checkpoint inhibitors along with radio-enhancing agents. Several unanswered questions remain concerning the exact mechanisms of action for nanomaterial-enhanced RT and for its combination with immune checkpoint inhibition and other immunostimulatory treatments in clinically relevant settings. The purpose of this article is to summarize key recent developments in this field and also highlight knowledge gaps that exist in this field. An improved mechanistic understanding will be critical for clinical translation of nanomaterials for advanced radio-immunotherapy. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Dhushyanth Viswanath
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Jeehun Park
- SOFT Foundry Institute, Seoul National University, Seoul, Republic of Korea
| | - Rahul Misra
- Analytical Sciences, Sanofi, Toronto, Ontario, Canada
| | - Vincenzo J Pizzuti
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sung-Ho Shin
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Junsang Doh
- SOFT Foundry Institute, Seoul National University, Seoul, Republic of Korea
- Department of Materials Science and Engineering, Institute of Engineering Research, BioMAX, Seoul National University, Seoul, Republic of Korea
| | - You-Yeon Won
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana, USA
- Purdue University Institute for Cancer Research, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
39
|
Batool A, Rashid W, Fatima K, Khan SU. Mechanisms of Cancer Resistance to Various Therapies. DRUG RESISTANCE IN CANCER: MECHANISMS AND STRATEGIES 2024:31-75. [DOI: 10.1007/978-981-97-1666-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
40
|
Jadhav T, Malik A, Kashif AW, Shelly D, Mishra PS, Baranwal AK, Sahu R. Study of the association of the known prognostic variables with EGFR expression in head and neck squamous cell carcinomas. INDIAN J PATHOL MICR 2024; 67:36-45. [PMID: 38358186 DOI: 10.4103/ijpm.ijpm_833_22] [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: 02/16/2024] Open
Abstract
Context : Head and neck squamous cell carcinomas (HNSCCs) are the sixth most frequent malignancy in the world. Epidermal growth factor receptors (EGFRs) are members of Erb B family of receptors. EGFR is known to act as a driver of tumorigenesis in various carcinomas. Over expression of EGFR in HNSCC is associated with poor prognosis and resistance to radiotherapy. It is a useful prognostic marker, marker for response to therapy, and also a therapeutic target. Aim To study the association of the known prognostic variables with EGFR expression in HNSCCs and to correlate it with the clinical outcome. Settings and Design Cross-sectional observational study. Materials and Methods A total of 170 patients of HNSCC were evaluated for EGFR expression and followed up for at least two years, with correlation of EGFR expression with various histopathological factors and their clinical outcome. Statistical Analysis Used : Chi-square test. Results The expression of EGFR in HNSCC in this study population was 88.82%. Statistical significance was noted between EGFR reactivity and age of the patient, its histological grade and perineural invasion. Statistical significance was also noted between EGFR reactivity and recurrence of malignancy as well as the site of recurrence. Conclusion EGFR expression in patients with HNSCC is a poor prognostic biomarker and has a comparatively lower survival outcome as compared to non-EGFR expressing HNSCC cases. Hence, it will be helpful for all those patients diagnosed with HNSCC to ideally undergo an additional EGFR immunohistochemical evaluation, which, in turn, will help the oncologists in management of the tumor with anti-EGFR therapy combined with radiotherapy, to obtain a better response and a survival outcome.
Collapse
Affiliation(s)
- Toyaja Jadhav
- Department of Laboratory Sciences, 12 Airforce Hospital, Akash Vihar, Gorakhpur, Uttar Pradesh, India
| | - Ajay Malik
- Yashoda Hospital and Research Centre, Nehru Nagar, Ghaziabad, Uttar Pradesh, India
| | - Ahmed Waheed Kashif
- Department of Pathology, Armed Forces Medical College, Wanowrie, Pune, Maharashtra, India
| | - Divya Shelly
- Department of Pathology, INHS Asvini, Mumbai, Maharashtra, India
| | - Prabhashankar S Mishra
- Department of Pathology, Army Research and Referral (R&R) Hospital, Dhaula Kuan, Delhi, India
| | - Ajay Kumar Baranwal
- Department of Blood Transfusion and Immunohematology, Armed Forces Medical College, Wanowrie, Pune, Maharashtra, India
| | - Rajesh Sahu
- Department of Community Medicine, Armed Forces Medical College, Wanowrie, Pune, Maharashtra, India
| |
Collapse
|
41
|
Telischi J, Nisenbaum E, Nicolli E. Carotid endarterectomy for the management of carotid stenosis occurring concurrently with head and neck cancer. Curr Opin Otolaryngol Head Neck Surg 2023; 31:452-456. [PMID: 37916904 DOI: 10.1097/moo.0000000000000928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
PURPOSE OF REVIEW Currently, most patients with concurrent head and neck cancer (HNC) and carotid stenosis (CS) are treated disjointedly for their oncologic and vascular lesions. The purpose of this review is to evaluate literature exploring a novel approach to these cases that poses several advantages, in which carotid endarterectomy (CEA) is performed simultaneously with surgical resection of HNC. RECENT FINDINGS Carotid stenosis is a common comorbidity of patients presenting with head and neck cancer as these pathologies have overlapping risk factors. Adjuvant oncologic therapy such as radiation therapy to the site of the lesion is known to increase development or progression of carotid stenosis. Performing simultaneous surgical management of CS and HNC decreases total procedures for the patient, provides a less challenging surgical field, and eliminates prioritization of treatment initiation for one pathology over the other. There has been limited reporting of simultaneous CEA with oncologic resection of HNC in the literature. However, of the 21 cases reviewed here, no perioperative strokes were reported with only one perioperative death from myocardial infarction. SUMMARY Available literature supports that simultaneous CEA with oncologic resection of HNC is safe and may offer several advantages, although larger studies are required.
Collapse
Affiliation(s)
- Julia Telischi
- Department of Otolaryngology, University of Miami Miller School of Medicine
| | - Eric Nisenbaum
- Department of Otolaryngology, University of Miami Miller School of Medicine
- Sylvester Comprehensive Cancer Center, Miami, Florida, USA
| | - Elizabeth Nicolli
- Department of Otolaryngology, University of Miami Miller School of Medicine
- Sylvester Comprehensive Cancer Center, Miami, Florida, USA
| |
Collapse
|
42
|
Medeiros MCD, The S, Bellile E, Russo N, Schmitd L, Danella E, Singh P, Banerjee R, Bassis C, Murphy GR, Sartor MA, Lombaert I, Schmidt TM, Eisbruch A, Murdoch-Kinch CA, Rozek L, Wolf GT, Li G, Chen GY, D'Silva NJ. Salivary microbiome changes distinguish response to chemoradiotherapy in patients with oral cancer. MICROBIOME 2023; 11:268. [PMID: 38037123 PMCID: PMC10687843 DOI: 10.1186/s40168-023-01677-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 09/26/2023] [Indexed: 12/02/2023]
Abstract
BACKGROUND Oral squamous cell carcinoma (SCC) is associated with oral microbial dysbiosis. In this unique study, we compared pre- to post-treatment salivary microbiome in patients with SCC by 16S rRNA gene sequencing and examined how microbiome changes correlated with the expression of an anti-microbial protein. RESULTS Treatment of SCC was associated with a reduction in overall bacterial richness and diversity. There were significant changes in the microbial community structure, including a decrease in the abundance of Porphyromonaceae and Prevotellaceae and an increase in Lactobacillaceae. There were also significant changes in the microbial community structure before and after treatment with chemoradiotherapy, but not with surgery alone. In patients treated with chemoradiotherapy alone, several bacterial populations were differentially abundant between responders and non-responders before and after therapy. Microbiome changes were associated with a change in the expression of DMBT1, an anti-microbial protein in human saliva. Additionally, we found that salivary DMBT1, which increases after treatment, could serve as a post-treatment salivary biomarker that links to microbial changes. Specifically, post-treatment increases in human salivary DMBT1 correlated with increased abundance of Gemella spp., Pasteurellaceae spp., Lactobacillus spp., and Oribacterium spp. This is the first longitudinal study to investigate treatment-associated changes (chemoradiotherapy and surgery) in the oral microbiome in patients with SCC along with changes in expression of an anti-microbial protein in saliva. CONCLUSIONS The composition of the oral microbiota may predict treatment responses; salivary DMBT1 may have a role in modulating the oral microbiome in patients with SCC. After completion of treatment, 6 months after diagnosis, patients had a less diverse and less rich oral microbiome. Leptotrichia was a highly prevalent bacteria genus associated with disease. Expression of DMBT1 was higher after treatment and associated with microbiome changes, the most prominent genus being Gemella Video Abstract.
Collapse
Affiliation(s)
- Marcell Costa de Medeiros
- Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave, Room G018, Ann Arbor, MI, 48109-1078, USA
| | - Stephanie The
- Cancer Data Science Shared Resource, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI, USA
| | - Emily Bellile
- Cancer Data Science Shared Resource, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI, USA
| | - Nickole Russo
- Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave, Room G018, Ann Arbor, MI, 48109-1078, USA
| | - Ligia Schmitd
- Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave, Room G018, Ann Arbor, MI, 48109-1078, USA
| | - Erika Danella
- Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave, Room G018, Ann Arbor, MI, 48109-1078, USA
| | - Priyanka Singh
- Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave, Room G018, Ann Arbor, MI, 48109-1078, USA
| | - Rajat Banerjee
- Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave, Room G018, Ann Arbor, MI, 48109-1078, USA
| | - Christine Bassis
- Internal Medicine, University of Michigan Medical School, 1500 East Medical Center Drive, Ann Arbor, MI, 331248109, USA
| | - George R Murphy
- Biologic and Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, 1011 N. University Ave, Ann Arbor, MI, USA
- Biointerfaces Institute, Ann Arbor, MI, USA
| | - Maureen A Sartor
- Computational Medicine and Bioinformatics, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI, USA
| | - Isabelle Lombaert
- Biologic and Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, 1011 N. University Ave, Ann Arbor, MI, USA
- Biointerfaces Institute, Ann Arbor, MI, USA
| | - Thomas M Schmidt
- Microbiology and Immunology, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI, USA
| | - Avi Eisbruch
- Radiation Oncology, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI, USA
| | - Carol Anne Murdoch-Kinch
- Oral Pathology, Medicine and Radiology, Indiana University School of Dentistry, 1011 North Michigan St, Indianapolis, IN, USA
| | - Laura Rozek
- Environmental Health Sciences, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI, USA
| | - Gregory T Wolf
- Otolaryngology, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI, USA
| | - Gen Li
- Biostatistics, University of Michigan School of Public Health, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI, USA
| | - Grace Y Chen
- Internal Medicine, University of Michigan Medical School, 1500 East Medical Center Drive, Ann Arbor, MI, 331248109, USA.
| | - Nisha J D'Silva
- Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave, Room G018, Ann Arbor, MI, 48109-1078, USA.
- Pathology, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI, USA.
- Rogel Cancer Center, Ann Arbor, MI, USA.
| |
Collapse
|
43
|
Jin Y, Wang Z, Huang S, Liu C, Wu X, Wang H. Identify and validate circadian regulators as potential prognostic markers and immune infiltrates in head and neck squamous cell carcinoma. Sci Rep 2023; 13:19939. [PMID: 37968308 PMCID: PMC10651996 DOI: 10.1038/s41598-023-46560-8] [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: 07/16/2023] [Accepted: 11/02/2023] [Indexed: 11/17/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a heterogeneity pathological malignant cancer with leading causes of morbidity and mortality. EGFR inhibitors, immune checkpoint inhibitors have become novel treatments. However, the mechanism still remained uncertain. Several studies have confirmed that the circadian rhythms induce multiple malignancies developing. We utilized multi-omics analysis to demonstrate the crosstalk between circadian clock genes and tumor microenvironment in HNSCC. Firstly, we performed the LASSO Cox regression analysis based on the 16 important clock genes. A 7-gene risk model was successfully established in TCGA and validated in GEO datasets. Next, CIBERSORT and ESTIMATE methods were performed to display the immune landscape of high risk and low risk groups, and the results showed that high abundance of mast cells activated, dendritic cells activated and neutrophils were positively correlated with poor OS. To further identify hub genes, Kaplan Meier plot was applied in all TCGA and GEO datasets and two hub genes (PER2, and PER3) were identified, especially PER3, which was found strongly associated with immune score, PDCD1, CD4 + and CD8 + T cells in HNSCCC. Moreover, to explore the innate mechanism of circadian-induced pathway, we constructed a circadian-related ceRNA regulatory network containing 34 lncRNAs, 3 miRNAs and 4 core circadian genes. In-vitro experiments also verified that Per2 or Per3 could suppressed the proliferation, migration and invasion of HNSC. This study unraveled the association between PER3 and prognosis in patients with HNSC and the innate mechanism remains to be elucidated.
Collapse
Affiliation(s)
- Yi Jin
- Department of Radiation Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- Key Laboratory of Translational Radiation Oncology, Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Zhanwang Wang
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Siwei Huang
- School of Humanities and Management, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Chang Liu
- Department of Nuclear Medicine, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Xiangwei Wu
- Department of Radiation Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- Key Laboratory of Translational Radiation Oncology, Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Hui Wang
- Department of Radiation Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.
- Key Laboratory of Translational Radiation Oncology, Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China.
| |
Collapse
|
44
|
Lu C, Sun Q, Guo Y, Han X, Zhang M, Liu J, Wang Y, Mou Y, Li Y, Song X. Construction and validation of a prognostic nine-gene signature associated with radiosensitivity in head and neck squamous cell carcinoma. Clin Transl Radiat Oncol 2023; 43:100686. [PMID: 37854672 PMCID: PMC10579965 DOI: 10.1016/j.ctro.2023.100686] [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: 07/27/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/20/2023] Open
Abstract
Background Radiotherapy is an effective treatment for head and neck squamous cell carcinoma (HNSCC), however how to predict the prognosis is not clear. Methods Here we collected 262 radiosensitivity-associated genes, screened and constructed a prognostic nine-gene risk model through univariate COX, lasso regression, stepwise regression and multivariate COX analysis for transcriptome and clinical information of HNSCC patients obtained from the cancer genome atlas (TCGA) and gene expression omnibus (GEO) databases. Results The reliability and robustness of the risk model were verified by receiver operating characteristic (ROC) curves, risk maps, and Kaplan-Meier (KM) curves analysis. Differences in immune cell infiltration and immune-related pathway enrichment between high-risk and low-risk subgroups were determined by multiple immune infiltration analyses. Meanwhile, the mutation map and the responses to immunotherapy were also differentiated by the prognostic nine-gene signature associated with radiosensitivity. These nine genes expression in HNSCC was verified in the Human Protein Atlas (HPA) database. After that, these nine genes expression was verified to be related to radiation resistance through in-vitro cell experiments. Conclusions All results showed that the nine-gene signature associated with radiosensitivity is a potential prognostic indicator for HNSCC patients after radiotherapy and provides potential gene targets for enhancing the efficacy of radiotherapy.
Collapse
Affiliation(s)
- Congxian Lu
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, China
| | - Qi Sun
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, China
| | - Ying Guo
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, China
| | - Xiao Han
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, China
| | - Mingjun Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, China
| | - Jiahui Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, China
| | - Yaqi Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, China
| | - Yakui Mou
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, China
| | - Yumei Li
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, China
| | - Xicheng Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, China
| |
Collapse
|
45
|
You P, Liu S, Li Q, Xie D, Yao L, Guo C, Guo Z, Wang T, Qiu H, Guo Y, Li J, Zhou H. Radiation-sensitive genetic prognostic model identifies individuals at risk for radiation resistance in head and neck squamous cell carcinoma. J Cancer Res Clin Oncol 2023; 149:15623-15640. [PMID: 37656244 DOI: 10.1007/s00432-023-05304-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 08/15/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND The advantages of radiotherapy for head and neck squamous cell carcinoma (HNSCC) depend on the radiation sensitivity of the patient. Here, we established and verified radiological factor-related gene signature and built a prognostic risk model to predict whether radiotherapy would be beneficial. METHODS Data from The Cancer Genome Atlas, Gene Expression Omnibus, and RadAtlas databases were subjected to LASSO regression, univariate COX regression, and multivariate COX regression analyses to integrate genomic and clinical information from patients with HNSCC. HNSCC radiation-related prognostic genes were identified, and patients classified into high- and low-risk groups, based on risk scores. Variations in radiation sensitivity according to immunological microenvironment, functional pathways, and immunotherapy response were investigated. Finally, the expression of HNSCC radiation-related genes was verified by qRT-PCR. RESULTS We built a clinical risk prediction model comprising a 15-gene signature and used it to divide patients into two groups based on their susceptibility to radiation: radiation-sensitive and radiation-resistant. Overall survival was significantly greater in the radiation-sensitive than the radiation-resistant group. Further, our model was an independent predictor of radiotherapy response, outperforming other clinical parameters, and could be combined with tumor mutational burden, to identify the target population with good predictive value for prognosis at 1, 2, and 3 years. Additionally, the radiation-resistant group was more vulnerable to low levels of immune infiltration, which are significantly associated with DNA damage repair, hypoxia, and cell cycle regulation. Tumor Immune Dysfunction and Exclusion scores also suggested that the resistant group would respond less favorably to immunotherapy. CONCLUSIONS Our prognostic model based on a radiation-related gene signature has potential for application as a tool for risk stratification of radiation therapy for patients with HNSCC, helping to identify candidates for radiation therapy and overcome radiation resistance.
Collapse
Affiliation(s)
- Peimeng You
- Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital, Nanchang, China
| | - Shengbo Liu
- Second Clinical College of Medicine, Southern Medical University, Guangzhou, China
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qiaxuan Li
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Daipeng Xie
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangzhou, China
| | - Lintong Yao
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Chenguang Guo
- Department of Radiation Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Zefeng Guo
- Department of Radiation Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Ting Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Hongrui Qiu
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Yangzhong Guo
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital, Nanchang, China
| | - Junyu Li
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital, Nanchang, China.
| | - Haiyu Zhou
- Nanchang University, Nanchang, China.
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Jiangxi Lung Cancer Institute, Nanchang, China.
| |
Collapse
|
46
|
Chang CL, Lin KC, Chen WM, Shia BC, Wu SY. Comparing the Oncologic Outcomes of Proton Therapy and Intensity-Modulated Radiation Therapy for Head and Neck Squamous Cell Carcinoma. Radiother Oncol 2023; 190:109971. [PMID: 39492511 DOI: 10.1016/j.radonc.2023.109971] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/12/2023] [Accepted: 10/25/2023] [Indexed: 11/05/2024]
Abstract
PURPOSE To compare the oncologic outcomes between proton therapy and intensity-modulated radiation therapy (IMRT) for head and neck squamous cell carcinoma (HNSCC) patients undergoing curative radiotherapy (RT). EXPERIMENTAL DESIGN We studied HNSCC patients who underwent curative-intent RT from 2015 to 2019, comparing the oncologic outcomes of proton therapy and IMRT. Our national retrospective HNSCC cohort study involved three institutes with proton therapy and 17 institutes (medical center levels) with IMRT in Taiwan. We utilized the Taiwan Cancer Registry Database to collect medical data for this study. We classified patients into two groups based on treatment method: Group 1 received IMRT, while Group 2 received proton therapy. 3:1 propensity score matching was performed to minimize the impact of potential confounders. Cox proportional hazards models were used to evaluate oncologic outcomes. RESULTS This study of 60,485 patients with HNSCC found that proton therapy was associated with better overall and cancer-specific survival and lower locoregional recurrence rates than IMRT. After matching, 982 patients were analyzed, with well-balanced factors. Proton therapy was a significant predictor of all-cause mortality, cancer-specific death, and locoregional recurrence (LRR). Patients who received proton therapy had significantly lower risks of all-cause mortality (adjusted hazard ratio, aHR=0.43), cancer-specific death (aHR=0.44), and LRR (aHR=0.61) than those who received IMRT. CONCLUSION Proton therapy is associated with superior outcomes in terms of overall survival, cancer-specific survival, and locoregional recurrence rates compared to IMRT in patients with HNSCC. These results provide valuable evidence for clinicians and patients in decision-making regarding the choice of radiation therapy for HNSCC.
Collapse
Affiliation(s)
- Chia-Lun Chang
- Department of Hemato-Oncology, Wan Fang Hospital, Taipei Medical University Taipei, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University Taipei, Taiwan
| | - Kuan-Chou Lin
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wan-Ming Chen
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, Taipei, Taiwan; Artificial Intelligence Development Center, Fu Jen Catholic University, Taipei, Taiwan
| | - Ben-Chang Shia
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, Taipei, Taiwan; Artificial Intelligence Development Center, Fu Jen Catholic University, Taipei, Taiwan
| | - Szu-Yuan Wu
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, Taipei, Taiwan; Artificial Intelligence Development Center, Fu Jen Catholic University, Taipei, Taiwan; Department of Food Nutrition and Health Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan; Division of Radiation Oncology, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan; Big Data Center, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan; Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung, Taiwan; Cancer Center, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan; Centers for Regional Anesthesia and Pain Medicine, Taipei Municipal Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Management, College of Management, Fo Guang University, Yilan, Taiwan.
| |
Collapse
|
47
|
Islam SS, Karakas B, Aboussekhra A, Noman ASM. KEAP1/NRF2 Mutations in Stem Cells Define an Aggressive Subset of Head and Neck Cancer Patients Who Have a Poor Prognosis, Lung Metastasis, and Therapeutic Failure. Cancers (Basel) 2023; 15:5006. [PMID: 37894373 PMCID: PMC10605399 DOI: 10.3390/cancers15205006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Mutations in Keap1/Nrf2 in head and neck cancer result in abnormal cell growth. Progenitor cells, bulk tumor cells, and head and neck cancer stem cells (HN-CSCs) may all harbor these mutations. Nevertheless, whether Keap1/Nrf2 mutations in HN-CSCs have an impact on clinical outcomes is unknown. Cancerous HN-CSCs and benign stem cells were obtained from freshly resected head and neck cancer patients (n = 50) via flow cytometry cell sorting and tested for Keap1/Nrf2 mutations. The existence of Keap1/Nrf2 mutations in HN-CSCs, as well as their correlations with tumor mutations, pathologic tumor stage, tumor histologic grades, lung metastasis, treatment outcomes, and the patient's age and conditions, are assessed at the last follow-up visit. Thirteen tumors were found to have Keap1/Nrf2 mutations in their HN-CSCs. More than half of the lung metastases and disease progression occurred in HN-CSCs with mutations. Patients whose tumors carried Keap1/Nrf2 mutations in their HN-CSCs had significantly shorter progression-free survival, overall survival, and time of treatment failure than their non-HN-CSC counterparts. These associations were partly driven by HN-CSCs, in which Keap1/Nrf2 mutations were overrepresented in fast progressors and associated with an increased risk of disease progression. Our findings suggest that molecular genotyping of HN-CSCs may facilitate personalized treatment strategies and assist in identifying patients who are likely to benefit from chemotherapy.
Collapse
Affiliation(s)
- Syed S. Islam
- Department Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
- Faculty of Medicine, Al-Faisal University, Riyadh 11533, Saudi Arabia
| | - Bedri Karakas
- 3 B & B Bio, 4 Professional Drive, Gaithersburg, MD 20879, USA;
| | - Abdelilah Aboussekhra
- Department Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Abu Shadat M. Noman
- Department Biochemistry and Molecular Biology, The University of Chittagong, Chittagong 4331, Bangladesh
| |
Collapse
|
48
|
Huang J, Meng Q, Liu R, Li H, Li Y, Yang Z, Wang Y, Wanyan C, Yang X, Wei J. The development of radioresistant oral squamous carcinoma cell lines and identification of radiotherapy-related biomarkers. Clin Transl Oncol 2023; 25:3006-3020. [PMID: 37029240 DOI: 10.1007/s12094-023-03169-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/21/2023] [Indexed: 04/09/2023]
Abstract
BACKGROUND In the treatment of oral squamous cell carcinoma (OSCC), radiation resistance remains an important obstacle to patient outcomes. Progress in understanding the molecular mechanisms of radioresistance has been limited by research models that do not fully recapitulate the biological features of solid tumors. In this study, we aimed to develop novel in vitro models to investigate the underlying basis of radioresistance in OSCC and to identify novel biomarkers. METHODS Parental OSCC cells (SCC9 and CAL27) were repeatedly exposed to ionizing radiation to develop isogenic radioresistant cell lines. We characterized the phenotypic differences between the parental and radioresistant cell lines. RNA sequencing was used to identify differentially expressed genes (DEGs), and bioinformatics analysis identified candidate molecules that may be related to OSCC radiotherapy. RESULTS Two isogenic radioresistant cell lines for OSCC were successfully established. The radioresistant cells displayed a radioresistant phenotype when compared to the parental cells. Two hundred and sixty DEGs were co-expressed in SCC9-RR and CAL27-RR, and thirty-eight DEGs were upregulated or downregulated in both cell lines. The associations between the overall survival (OS) of OSCC patients and the identified genes were analyzed using data from the Cancer Genome Atlas (TCGA) database. A total of six candidate genes (KCNJ2, CLEC18C, P3H3, PIK3R3, SERPINE1, and TMC8) were closely associated with prognosis. CONCLUSION This study demonstrated the utility of constructing isogenic cell models to investigate the molecular changes associated with radioresistance. Six genes were identified based on the data from the radioresistant cells that may be potential targets in the treatment of OSCC.
Collapse
Affiliation(s)
- Junhong Huang
- College of Life Science, Northwest University, Xi'an, 710069, China
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Qingzhe Meng
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
- School of Stomatology, Heilongjiang Key Lab of Oral Biomedicine Materials and Clinical Application & Experimental Center for Stomatology Engineering, Jiamusi University, Jiamusi, 154000, China
| | - Rong Liu
- College of Life Science, Northwest University, Xi'an, 710069, China
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Huan Li
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yahui Li
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Zihui Yang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yan Wang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Chaojie Wanyan
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Xinjie Yang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jianhua Wei
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
49
|
Green V, Baldwin L, England J, Marshall G, Frost L, Moore C, Greenman J. Head and Neck Squamous Cell Carcinoma Biopsies Maintained Ex Vivo on a Perfusion Device Show Gene Changes with Time and Clinically Relevant Doses of Irradiation. Cancers (Basel) 2023; 15:4575. [PMID: 37760543 PMCID: PMC10527562 DOI: 10.3390/cancers15184575] [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: 08/23/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Advancements in 3-Dimensional (3D) culture models for studying disease have increased significantly over the last two decades, but fully understanding how these models represent in vivo still requires further investigation. The current study investigated differences in gene expression between a baseline sample and that maintained on a tissue-on-chip perfusion device for up to 96 h, with and without clinically-relevant doses of irradiation, to allow differentiation of model and treatment effects. Tumour tissue samples from 7 Head and Neck Squamous Cell Carcinomas (HNSCC) patients were sub-divided and either fixed immediately upon excision or maintained in a tissue-on-chip device for 48 and 96 h, with or without 2 Gray (Gy) or 10 Gy irradiation. Gene expression was measured using an nCounter® PanCancer Progression Panel. Differentially expressed genes between pre- and post-ex vivo culture, and control and irradiated samples were identified using nSolver software (version 4.0). The secretome from the tumour-on-chip was analysed for the presence of cytokines using a Proteome Profiler™ platform. Significant numbers of genes both increased (n = 6 and 64) and decreased (n = 18 and 58) in expression in the tissue maintained on-chip for 48 and 96 h, respectively, compared to fresh tissue; however, the irradiation schedule chosen did not induce significant changes in gene expression or cytokine secretion. Although HNSCC tissue maintained ex vivo shows a decrease in a large proportion of altered genes, 25% and 53% (48 and 96 h) do show increased expression, suggesting that the tissue remains functional. Irradiation of tumour tissue-on-chip needs to be conducted for longer time periods for specific gene changes to be observed, but we have shown, for the first time, the feasibility of using this perfusion platform for studying the genomic response of HNSCC tissue biopsies.
Collapse
Affiliation(s)
- Victoria Green
- Centre for Biomedicine, Hull York Medical School, Faculty of Health Sciences, University of Hull, Hull HU6 7RX, UK; (L.B.); (J.G.)
| | - Lydia Baldwin
- Centre for Biomedicine, Hull York Medical School, Faculty of Health Sciences, University of Hull, Hull HU6 7RX, UK; (L.B.); (J.G.)
| | - James England
- Department of Otorhinolaryngology, Head and Neck Surgery, Hull University Teaching Hospitals NHS Trust Hull, Hull HU16 5JQ, UK;
| | - Gayle Marshall
- Medicines Discovery Catapult Ltd., Alderley Park, Alderley Edge, Cheshire SK10 4TG, UK; (G.M.); (L.F.)
| | - Lucy Frost
- Medicines Discovery Catapult Ltd., Alderley Park, Alderley Edge, Cheshire SK10 4TG, UK; (G.M.); (L.F.)
| | - Craig Moore
- Medical Physics Service, Hull University Teaching Hospitals NHS Trust Hull, Hull HU16 5JQ, UK;
| | - John Greenman
- Centre for Biomedicine, Hull York Medical School, Faculty of Health Sciences, University of Hull, Hull HU6 7RX, UK; (L.B.); (J.G.)
| |
Collapse
|
50
|
Ricco N, Kron SJ. Statins in Cancer Prevention and Therapy. Cancers (Basel) 2023; 15:3948. [PMID: 37568764 PMCID: PMC10417177 DOI: 10.3390/cancers15153948] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/29/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023] Open
Abstract
Statins, a class of HMG-CoA reductase inhibitors best known for their cholesterol-reducing and cardiovascular protective activity, have also demonstrated promise in cancer prevention and treatment. This review focuses on their potential applications in head and neck cancer (HNC), a common malignancy for which established treatment often fails despite incurring debilitating adverse effects. Preclinical and clinical studies have suggested that statins may enhance HNC sensitivity to radiation and other conventional therapies while protecting normal tissue, but the underlying mechanisms remain poorly defined, likely involving both cholesterol-dependent and -independent effects on diverse cancer-related pathways. This review brings together recent discoveries concerning the anticancer activity of statins relevant to HNC, highlighting their anti-inflammatory activity and impacts on DNA-damage response. We also explore molecular targets and mechanisms and discuss the potential to integrate statins into conventional HNC treatment regimens to improve patient outcomes.
Collapse
Affiliation(s)
- Natalia Ricco
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08195 Barcelona, Spain;
| | - Stephen J. Kron
- Department of Molecular Genetics and Cell Biology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|