1
|
Lai J, Yang H, Chen J, Chen S, Chen X. Predicting radiotherapy efficacy and prognosis in tongue squamous cell carcinoma through an in-depth analysis of a radiosensitivity gene signature. Front Oncol 2024; 14:1334747. [PMID: 39252950 PMCID: PMC11381225 DOI: 10.3389/fonc.2024.1334747] [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: 11/08/2023] [Accepted: 08/07/2024] [Indexed: 09/11/2024] Open
Abstract
Background Tongue squamous cell carcinoma (TSCC) is a prevalent tumor that affects many people worldwide. Radiotherapy is a common treatment option, but its efficacy varies greatly. This study seeks to validate the identified gene signature associated with radiosensitivity in TSCC, and its potential in predicting radiotherapy response and prognosis. Methods We analyzed 122 TSCC patients from TCGA database using the radiosensitivity signature and classified them into radiosensitive (RS) and radioresistant (RR) groups. Immune infiltration analysis methods were applied to investigate the immune status between different subgroups. Immunophenotype Score (IPS) and pRRophetic algorithm were employed to estimate the efficiency of treatment. A radioresistant TSCC cell line was established by gradually increasing radiation doses. Cell radiosensitivity was evaluated using the CCK-8 and colony formation assays. The expression of radiosensitivity-related genes was validated by qRT-PCR. Results Our study validated the predictive capacity of a previously identified "31-gene signature" in the TCGA-TSCC cohort, which effectively stratified patients into RS and RR groups. We observed that the RS group exhibited superior overall survival and progression-free survival rates relative to the RR group when treated with radiotherapy. The RS group was significantly enriched in most immune-related hallmark pathways, and may therefore benefit from immune checkpoint inhibitors. However, the RS group displayed lower sensitivity to first-line chemotherapy. A radioresistant TSCC cell line (CAL-27R) exhibited increased clonogenic potential and cell viability following irradiation, accompanied by downregulation of three radiosensitivity-related genes compared to its parental non-resistant cell (CAL-27). In addition, we constructed and validated a radiosensitivity-related prognostic index (PI) using 4 radiosensitivity-related genes associated with TSCC prognosis. Conclusion We assessed the ability of the radiosensitivity gene signature to predict outcomes in TSCC patients. our research provided valuable insights into the molecular pathways associated with radiosensitivity in TSCC and offered clinicians a practical tool to predict patient radiotherapy effectiveness and prognosis.
Collapse
Affiliation(s)
- Jinzhi Lai
- Department of Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Hainan Yang
- Department of Ultrasound, First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Junjun Chen
- National Health Commission (NHC) Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Jiangxi Cancer Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Shoubo Chen
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Xiaofang Chen
- Department of Otolaryngology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
2
|
Choi SY, Shim J, Gu DE, Kim SY, Kim HJ, Shin DY, Chung MK. Clonal evolution of long-term expanding head and neck cancer organoid: Impact on treatment response for personalized therapeutic screening. Oral Oncol 2023; 146:106571. [PMID: 37741019 DOI: 10.1016/j.oraloncology.2023.106571] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/27/2023] [Accepted: 09/16/2023] [Indexed: 09/25/2023]
Abstract
OBJECTIVES In biobanking based on patient-derived organoids (PDO), the genetic stability of organoid lines is critical for the clinical relevance of PDO with parental tumors. However, data on mutational heterogeneity and clonal evolution of PDO and their effects on treatment response are insufficient. METHODS To investigate whether head and neck cancer organoids (HNCOs) could maintain the genetic characteristics of their original tumors and elucidate the clonal evolution process during a long-term passage, we performed targeted sequencing, covering 377 cancer-related genes and adopted a sub-clonal fraction model. To explore therapeutic response variability between an early and late passage (>passage 6), we generated dose-response curves for drugs and radiation using two HNCO lines. RESULTS Using 3D ex vivo organoid culture protocol, we successfully established 27 HNCOs from 39 patients with an overall success rate of 70% (27/39). Their mutational profiles were highly concordant, with three of the HNCOs analyzed showing greater than 70% concordance. Only one HNCO displayed less than 50% concordance. However, many of these organoid lines displayed clonal evolution during serial passaging, although major cancer driver genes and VAF distributions were shared between early and later passages. We also found that all late passages of HNCOs tended to be more sensitive to radiation than early passages, similar to drug response results. CONCLUSIONS We report the establishment of HNCO lines derived from 27 patients and demonstrate their genetic concordance with corresponding parental tumors. Furthermore, we show serial changes in mutational profiles of HNCO along with long passage culture and the impact of these clonal evolutions on response to radiotherapy.
Collapse
Affiliation(s)
- Sung Yong Choi
- Department of Otorhinolaryngology-Head and Neck Surgery, National Cancer Center, Goyang, Republic of Korea
| | - Joonho Shim
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Do-Eon Gu
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Soo Yoon Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Hye Jin Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Da-Yong Shin
- Department of Otorhinolaryngology-Head and Neck Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Man Ki Chung
- Department of Otorhinolaryngology-Head and Neck Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea.
| |
Collapse
|
3
|
Corti A, De Cecco L, Cavalieri S, Lenoci D, Pistore F, Calareso G, Mattavelli D, de Graaf P, Leemans CR, Brakenhoff RH, Ravanelli M, Poli T, Licitra L, Corino V, Mainardi L. MRI-based radiomic prognostic signature for locally advanced oral cavity squamous cell carcinoma: development, testing and comparison with genomic prognostic signatures. Biomark Res 2023; 11:69. [PMID: 37455307 DOI: 10.1186/s40364-023-00494-5] [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: 03/03/2023] [Accepted: 05/03/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND . At present, the prognostic prediction in advanced oral cavity squamous cell carcinoma (OCSCC) is based on the tumor-node-metastasis (TNM) staging system, and the most used imaging modality in these patients is magnetic resonance image (MRI). With the aim to improve the prediction, we developed an MRI-based radiomic signature as a prognostic marker for overall survival (OS) in OCSCC patients and compared it with published gene expression signatures for prognosis of OS in head and neck cancer patients, replicated herein on our OCSCC dataset. METHODS For each patient, 1072 radiomic features were extracted from T1 and T2-weighted MRI (T1w and T2w). Features selection was performed, and an optimal set of five of them was used to fit a Cox proportional hazard regression model for OS. The radiomic signature was developed on a multi-centric locally advanced OCSCC retrospective dataset (n = 123) and validated on a prospective cohort (n = 108). RESULTS The performance of the signature was evaluated in terms of C-index (0.68 (IQR 0.66-0.70)), hazard ratio (HR 2.64 (95% CI 1.62-4.31)), and high/low risk group stratification (log-rank p < 0.001, Kaplan-Meier curves). When tested on a multi-centric prospective cohort (n = 108), the signature had a C-index of 0.62 (IQR 0.58-0.64) and outperformed the clinical and pathologic TNM stage and six out of seven gene expression prognostic signatures. In addition, the significant difference of the radiomic signature between stages III and IVa/b in patients receiving surgery suggests a potential association of MRI features with the pathologic stage. CONCLUSIONS Overall, the present study suggests that MRI signatures, containing non-invasive and cost-effective remarkable information, could be exploited as prognostic tools.
Collapse
Affiliation(s)
- Anna Corti
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy.
| | - Loris De Cecco
- Integrated Biology of Rare Tumors, Department of Research, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Stefano Cavalieri
- Head and Neck Medical Oncology Department, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
- Department of Oncology and Hemato-Oncology, Università degli studi di Milano, Milan, Italy
| | - Deborah Lenoci
- Integrated Biology of Rare Tumors, Department of Research, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Federico Pistore
- Head and Neck Medical Oncology Department, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Giuseppina Calareso
- Radiology Department, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Davide Mattavelli
- Unit of Otorhinolaryngology-Head and Neck Surgery, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, ASST Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Pim de Graaf
- Amsterdam UMC location Vrije Universiteit, Radiology and Nuclear Medicine, de Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - C René Leemans
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit, Otolaryngology-Head and Neck Surgery, de Boelelaan 1117, Amsterdam, The Netherlands
| | - Ruud H Brakenhoff
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit, Otolaryngology-Head and Neck Surgery, de Boelelaan 1117, Amsterdam, The Netherlands
| | - Marco Ravanelli
- Unit of Radiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, ASST Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Tito Poli
- Maxillo-Facial Surgery Division, Head and Neck Department, University Hospital of Parma, Parma, Italy
| | - Lisa Licitra
- Head and Neck Medical Oncology Department, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
- Department of Oncology and Hemato-Oncology, Università degli studi di Milano, Milan, Italy
| | - Valentina Corino
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
- Cardiotech Lab, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Luca Mainardi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| |
Collapse
|
4
|
Zeng Z, Luo M, Li Y, Li J, Huang Z, Zeng Y, Yuan Y, Wang M, Liu Y, Gong Y, Xie C. Prediction of radiosensitivity and radiocurability using a novel supervised artificial neural network. BMC Cancer 2022; 22:1243. [PMID: 36451111 PMCID: PMC9713966 DOI: 10.1186/s12885-022-10339-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/21/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Radiotherapy has been widely used to treat various cancers, but its efficacy depends on the individual involved. Traditional gene-based machine-learning models have been widely used to predict radiosensitivity. However, there is still a lack of emerging powerful models, artificial neural networks (ANN), in the practice of gene-based radiosensitivity prediction. In addition, ANN may overfit and learn biologically irrelevant features. METHODS We developed a novel ANN with Selective Connection based on Gene Patterns (namely ANN-SCGP) to predict radiosensitivity and radiocurability. We creatively used gene patterns (gene similarity or gene interaction information) to control the "on-off" of the first layer of weights, enabling the low-dimensional features to learn the gene pattern information. ANN-SCGP was trained and tested in 82 cell lines and 1,101 patients from the 11 pan-cancer cohorts. RESULTS For survival fraction at 2 Gy, the root mean squared errors (RMSE) of prediction in ANN-SCGP was the smallest among all algorithms (mean RMSE: 0.1587-0.1654). For radiocurability, ANN-SCGP achieved the first and second largest C-index in the 12/20 and 4/20 tests, respectively. The low dimensional output of ANN-SCGP reproduced the patterns of gene similarity. Moreover, the pan-cancer analysis indicated that immune signals and DNA damage responses were associated with radiocurability. CONCLUSIONS As a model including gene pattern information, ANN-SCGP had superior prediction abilities than traditional models. Our work provided novel insights into radiosensitivity and radiocurability.
Collapse
Affiliation(s)
- Zihang Zeng
- grid.413247.70000 0004 1808 0969Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071 Hubei China
| | - Maoling Luo
- grid.413247.70000 0004 1808 0969Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071 Hubei China
| | - Yangyi Li
- grid.413247.70000 0004 1808 0969Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071 Hubei China
| | - Jiali Li
- grid.413247.70000 0004 1808 0969Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071 Hubei China
| | - Zhengrong Huang
- grid.413247.70000 0004 1808 0969Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071 Hubei China ,grid.413247.70000 0004 1808 0969Department of Biological Repositories, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071 Hubei China
| | - Yuxin Zeng
- grid.413247.70000 0004 1808 0969Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071 Hubei China
| | - Yu Yuan
- grid.413247.70000 0004 1808 0969Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071 Hubei China
| | - Mengqin Wang
- grid.413247.70000 0004 1808 0969Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071 Hubei China
| | - Yuying Liu
- grid.413247.70000 0004 1808 0969Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071 Hubei China
| | - Yan Gong
- grid.413247.70000 0004 1808 0969Department of Biological Repositories, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071 Hubei China ,grid.413247.70000 0004 1808 0969Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071 Hubei China
| | - Conghua Xie
- grid.413247.70000 0004 1808 0969Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071 Hubei China ,grid.413247.70000 0004 1808 0969Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, 430071 Hubei China ,grid.413247.70000 0004 1808 0969Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071 Hubei China
| |
Collapse
|
5
|
A novel 3D pillar/well array platform using patient-derived head and neck tumor to predict the individual radioresponse. Transl Oncol 2022; 24:101483. [PMID: 35850059 PMCID: PMC9294182 DOI: 10.1016/j.tranon.2022.101483] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/24/2022] [Accepted: 07/06/2022] [Indexed: 12/24/2022] Open
Abstract
Radiotherapy is a critical modality in head and neck cancer treatment. A novel 3D pillar/well array platform provides the individual radioresponse biomarker, RTauc. Poor and good radioresponse group by RTauc correlates with other clinical features. RTauc shows potential for radioresponse biomarker, useful in clinical decision-making.
Predicting individual radiotherapy (RT) response is valuable in managing head and neck squamous cell carcinoma (HNSCC). We assessed the feasibility of our novel 3D culture platform to measure radioresponse using patient-derived cells (PDCs) from HNSCC patients. Cells from the FaDu line and tumor samples from 39 HNSCC patients were cultivated serially in MatrigelTM on a 3D pillar/well array culture system. The 3D tumor models were exposed to 0 to 8 Gy of radiation dose, and the radioresponse index (RTauc, area under the dose-response curve) was measured quantitatively with Calcein AM staining of live tumor cells. Calcein AM fluorescence showed reduced density and the number of FaDu colonies as radiation increased, implying a dose-dependent effect on cell viability in the 3D pillar/well culture system. 3D tumor models using PDCs were established successfully from 39 HNSCC patient tumor samples, maintaining original genomic and pathological characteristics. These 3D tumor models were exposed to ionizing radiation on a 3D pillar/well array, with a mean period of 12 days from tumor harvest to the measurement of RTauc. The RTauc of all PDCs varied from 3.5 to 9.4, and the lower 40th percentile (Z-score = -0.26) was considered a good radioresponse group with a threshold RTauc of 4.6. The good radioresponse group showed fewer adverse features than others. As of the last follow-up, recurrence-free survival was better in the good radioresponse group (p = 0.037). 3D pillar/well array platforms using PDC could rapidly quantify radioresponse index in patients with HNSCC, showing potential as a novel prognosticator.
Collapse
|
6
|
Zhu G, Yang K, Xu C, Feng R, Li W, Ma J. Development of a prediction model for radiotherapy response among patients with head and neck squamous cell carcinoma based on the tumor immune microenvironment and hypoxia signature. Cancer Med 2022; 11:4673-4687. [PMID: 35505641 PMCID: PMC9741991 DOI: 10.1002/cam4.4791] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/26/2022] [Accepted: 04/18/2022] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION The immune system and hypoxia are major factors influencing radiosensitivity in patients with different cancer types. This study aimed at developing a model to predict radiotherapy response in patients with head and neck squamous cell carcinoma (HNSCC) based on the tumor immune microenvironment and hypoxia signature. MATERIALS AND METHODS We first evaluated the hypoxia status and tumor immune microenvironment in the Cancer Genome Atlas (TCGA) cohort by using transcriptomic data. Differentially expressed genes (DEGs) were identified between the "high immunity and low hypoxia" and "low immunity and high hypoxia" groups and those DEGs significantly associated with disease-specific survival in the univariate Cox regression analysis were selected as the prognostic DEGs. We selected the immune hypoxia-related genes (IHRGs) by intersecting prognostic DEGs with immune and hypoxia gene sets. We used the IHRGs to train a multivariate Cox regression model in the TCGA cohort, based on which we calculated the IHRG prognostic index (IHRGPI) for each patient and validated its efficacy in predicting radiotherapy response in the Gene Expression Omnibus cohorts. Furthermore, we explored potential mechanisms and effective combinational treatment strategies for different IHRGPI groups. RESULTS Five IHRGs were used to construct the IHRGPI, which was used to dichotomize the cohorts. The patients with lower IHRGPI showed a better radiotherapy response across different cohorts and endpoints, including overall survival, progression-free survival, and recurrence-free survival (p < 0.05). Patients with higher IHRGPI showed greater hypoxia and lesser immune cell infiltration. A lower IHRGPI indicated a better immunotherapy response, while a higher IHRGPI indicated a better chemotherapy response. CONCLUSIONS IHRGPI is promising for predicting radiotherapy response and guiding combinational treatment strategies in patients with HNSCC.
Collapse
Affiliation(s)
- Guang‐Li Zhu
- Department of Radiation OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangzhouP. R. China
| | - Kai‐Bin Yang
- Department of Radiation OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangzhouP. R. China
| | - Cheng Xu
- Department of Radiation OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangzhouP. R. China
| | - Rui‐Jia Feng
- Department of Radiation OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangzhouP. R. China
| | - Wen‐Fei Li
- Department of Radiation OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangzhouP. R. China
| | - Jun Ma
- Department of Radiation OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangzhouP. R. China
| |
Collapse
|
7
|
Byrne NM, Tambe P, Coulter JA. Radiation Response in the Tumour Microenvironment: Predictive Biomarkers and Future Perspectives. J Pers Med 2021; 11:jpm11010053. [PMID: 33467153 PMCID: PMC7830490 DOI: 10.3390/jpm11010053] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy (RT) is a primary treatment modality for a number of cancers, offering potentially curative outcomes. Despite its success, tumour cells can become resistant to RT, leading to disease recurrence. Components of the tumour microenvironment (TME) likely play an integral role in managing RT success or failure including infiltrating immune cells, the tumour vasculature and stroma. Furthermore, genomic profiling of the TME could identify predictive biomarkers or gene signatures indicative of RT response. In this review, we will discuss proposed mechanisms of radioresistance within the TME, biomarkers that may predict RT outcomes, and future perspectives on radiation treatment in the era of personalised medicine.
Collapse
|