1
|
Rifi AL, Raets C, Dufait I, De Ridder M, Barbé K. Integrating radiomics and immunology: Non-invasive assessment of intratumoral CD8 + T cell levels in murine models. Comput Biol Med 2025; 190:110061. [PMID: 40138967 DOI: 10.1016/j.compbiomed.2025.110061] [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: 11/29/2024] [Revised: 03/03/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Radiomics has emerged as a promising tool for extracting quantitative features from medical images, enabling deeper insights into tumor phenotypes and microenvironmental heterogeneity. Nonetheless, the clinical application of these features is impeded by their inherent non-biological interpretability. It is, therefore, critically important to unravel the biological meaning of these radiomic features. This study aims to address this gap by integrating radiomics with immunology to establish a direct link between imaging biomarkers and immune dynamics. Using murine cancer models, we manipulated the tumor microenvironment with monoclonal antibodies to modulate CD8+ T cell levels, a critical determinant of antitumoral immunity. Tumors were imaged, segmented, and analyzed for radiomic feature extraction, followed by immune cell quantification. Through exploratory factor analysis, we identified a radiomic signature that reflects intratumoral CD8+ T cell levels, establishing a novel non-invasive biomarker. This work enhances the explainability of radiomic features and supports their potential role in immune profiling within preclinical models. Future validation in human imaging datasets is necessary to determine their translational relevance for precision oncology.
Collapse
Affiliation(s)
- Amir L Rifi
- Biostatistics and Medical Informatics (BISI) Research Group, Vrije Universiteit Brussels, Belgium; Department of Radiotherapy Research Group, UZ Brussels, Belgium.
| | - Camille Raets
- Biostatistics and Medical Informatics (BISI) Research Group, Vrije Universiteit Brussels, Belgium; Department of Radiotherapy Research Group, UZ Brussels, Belgium
| | - Inès Dufait
- Department of Radiotherapy Research Group, UZ Brussels, Belgium
| | - Mark De Ridder
- Department of Radiotherapy Research Group, UZ Brussels, Belgium
| | - Kurt Barbé
- Biostatistics and Medical Informatics (BISI) Research Group, Vrije Universiteit Brussels, Belgium; Department of Radiotherapy Research Group, UZ Brussels, Belgium
| |
Collapse
|
2
|
Bernatowicz K, Amat R, Prior O, Frigola J, Ligero M, Grussu F, Zatse C, Serna G, Nuciforo P, Toledo R, Escobar M, Garralda E, Felip E, Perez-Lopez R. Radiomics signature for dynamic monitoring of tumor inflamed microenvironment and immunotherapy response prediction. J Immunother Cancer 2025; 13:e009140. [PMID: 39800381 PMCID: PMC11749429 DOI: 10.1136/jitc-2024-009140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 11/14/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND The efficacy of immune checkpoint inhibitors (ICIs) depends on the tumor immune microenvironment (TIME), with a preference for a T cell-inflamed TIME. However, challenges in tissue-based assessments via biopsies have triggered the exploration of non-invasive alternatives, such as radiomics, to comprehensively evaluate TIME across diverse cancers. To address these challenges, we develop an ICI response signature by integrating radiomics with T cell-inflamed gene-expression profiles. METHODS We conducted a pan-cancer investigation into the utility of radiomics for TIME assessment, including 1360 tumors from 428 patients. Leveraging contrast-enhanced CT images, we characterized TIME through RNA gene expression analysis, using the T cell-inflamed gene expression signature. Subsequently, a pan-cancer CT-radiomic signature predicting inflamed TIME (CT-TIME) was developed and externally validated. Machine learning was employed to select robust radiomic features and predict inflamed TIME. The study also integrated independent cohorts with longitudinal CT images, baseline biopsies, and comprehensive immunohistochemistry panel evaluation to assess the pan-cancer biological associations, spatiotemporal landscape and clinical utility of the CT-TIME. RESULTS The CT-TIME signature, comprising four radiomic features linked to a T-cell inflamed microenvironment, demonstrated robust performance with AUCs (95% CI) of 0.85 (0.73 to 0.96) (training) and 0.78 (0.65 to 0.92) (external validation). CT-TIME scores exhibited positive correlations with CD3, CD8, and CD163 expression. Intrapatient analysis revealed considerable heterogeneity in TIME between tumors, which could not be assessed using biopsies. Evaluation of aggregated per-patient CT-TIME scores highlighted its promising clinical utility for dynamically assessing the immune microenvironment and predicting immunotherapy response across diverse scenarios in advanced cancer. Despite demonstrating progression disease at the first follow-up, patients within the inflamed status group, identified by CT-TIME, exhibited significantly prolonged progression-free survival (PFS), with some surpassing 5 months, suggesting a potential phenomenon of pseudoprogression. Cox models using aggregated CT-TIME scores from baseline images revealed a statistically significant reduction in the risk of PFS in the pan-cancer cohort (HR 0.62, 95% CI 0.44 to 0.88, p=0.007), and Kaplan-Meier analysis further confirmed substantial differences in PFS between patients with inflamed and uninflamed status (log-rank test p=0.009). CONCLUSIONS The signature holds promise for impacting clinical decision-making, pan-cancer patient stratification, and treatment outcomes in immune checkpoint therapies.
Collapse
Affiliation(s)
| | - Ramon Amat
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Olivia Prior
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Joan Frigola
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Marta Ligero
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | | | - Garazi Serna
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | | | | | - Elena Garralda
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Vall d'Hebron University Hospital, Barcelona, Spain
| | - Enriqueta Felip
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Vall d'Hebron University Hospital, Barcelona, Spain
| | - Raquel Perez-Lopez
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Vall d'Hebron University Hospital, Barcelona, Spain
| |
Collapse
|
3
|
Geng X, Zhang Y, Li Y, Cai Y, Liu J, Geng T, Meng X, Hao F. Radiomics-clinical nomogram for preoperative lymph node metastasis prediction in esophageal carcinoma. Br J Radiol 2024; 97:652-659. [PMID: 38268475 PMCID: PMC11027331 DOI: 10.1093/bjr/tqae009] [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: 07/14/2023] [Revised: 11/10/2023] [Accepted: 12/18/2023] [Indexed: 01/26/2024] Open
Abstract
OBJECTIVES This research aimed to develop a radiomics-clinical nomogram based on enhanced thin-section CT radiomics and clinical features for the purpose of predicting the presence or absence of metastasis in lymph nodes among patients with resectable esophageal squamous cell carcinoma (ESCC). METHODS This study examined the data of 256 patients with ESCC, including 140 cases with lymph node metastasis. Clinical information was gathered for each case, and radiomics features were derived from thin-section contrast-enhanced CT with the help of a 3D slicer. To validate risk factors that are independent of the clinical and radiomics models, least absolute shrinkage and selection operator logistic regression analysis was used. A nomogram pattern was constructed based on the radiomics features and clinical characteristics. The receiver operating characteristic curve and Brier Score were used to evaluate the model's discriminatory ability, the calibration plot to evaluate the model's calibration, and the decision curve analysis to evaluate the model's clinical utility. The confusion matrix was used to evaluate the applicability of the model. To evaluate the efficacy of the model, 1000 rounds of 5-fold cross-validation were conducted. RESULTS The clinical model identified esophageal wall thickness and clinical T (cT) stage as independent risk factors, whereas the radiomics pattern was built based on 4 radiomics features chosen at random. Area under the curve (AUC) values of 0.684 and 0.701 are observed for the radiomics approach and clinical model, respectively. The AUC of nomogram combining radiomics and clinical features was 0.711. The calibration plot showed good agreement between the incidence of lymph node metastasis predicted by the nomogram and the actual probability of occurrence. The nomogram model displayed acceptable levels of performance. After 1000 rounds of 5-fold cross-validation, the AUC and Brier score had median values of 0.702 (IQR: 0.65, 7.49) and 0.21 (IQR: 0.20, 0.23), respectively. High-risk patients (risk point >110) were found to have an increased risk of lymph node metastasis [odds ratio (OR) = 5.15, 95% CI, 2.95-8.99] based on the risk categorization. CONCLUSION A successful preoperative prediction performance for metastasis to the lymph nodes among patients with ESCC was demonstrated by the nomogram that incorporated CT radiomics, wall thickness, and cT stage. ADVANCES IN KNOWLEDGE This study demonstrates a novel radiomics-clinical nomogram for lymph node metastasis prediction in ESCC, which helps physicians determine lymph node status preoperatively.
Collapse
Affiliation(s)
- Xiaotao Geng
- Shandong University Cancer Center, Shandong University, 440 Jiyan Road, Jinan, 250117, China
- Department of Radiation Oncology, Weifang People’s Hospital, 151 Guangwen Street, Weifang, 261000, China
| | - Yaping Zhang
- Department of Radiology, Weifang People’s Hospital, 151 Guangwen Street, Weifang, 261000, China
| | - Yang Li
- Department of Radiation Oncology, Weifang People’s Hospital, 151 Guangwen Street, Weifang, 261000, China
| | - Yuanyuan Cai
- Department of Radiation Oncology, Weifang People’s Hospital, 151 Guangwen Street, Weifang, 261000, China
| | - Jie Liu
- Department of Radiation Oncology, Weifang People’s Hospital, 151 Guangwen Street, Weifang, 261000, China
| | - Tianxiang Geng
- Department of Biomaterials, Faculty of Dentistry, University of Oslo, Oslo, 0455, Norway
| | - Xiangdi Meng
- Department of Radiation Oncology, Weifang People’s Hospital, 151 Guangwen Street, Weifang, 261000, China
| | - Furong Hao
- Department of Radiation Oncology, Weifang People’s Hospital, 151 Guangwen Street, Weifang, 261000, China
| |
Collapse
|
4
|
Ligero M, Gielen B, Navarro V, Cresta Morgado P, Prior O, Dienstmann R, Nuciforo P, Trebeschi S, Beets-Tan R, Sala E, Garralda E, Perez-Lopez R. A whirl of radiomics-based biomarkers in cancer immunotherapy, why is large scale validation still lacking? NPJ Precis Oncol 2024; 8:42. [PMID: 38383736 PMCID: PMC10881558 DOI: 10.1038/s41698-024-00534-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/26/2024] [Indexed: 02/23/2024] Open
Abstract
The search for understanding immunotherapy response has sparked interest in diverse areas of oncology, with artificial intelligence (AI) and radiomics emerging as promising tools, capable of gathering large amounts of information to identify suitable patients for treatment. The application of AI in radiology has grown, driven by the hypothesis that radiology images capture tumor phenotypes and thus could provide valuable insights into immunotherapy response likelihood. However, despite the rapid growth of studies, no algorithms in the field have reached clinical implementation, mainly due to the lack of standardized methods, hampering study comparisons and reproducibility across different datasets. In this review, we performed a comprehensive assessment of published data to identify sources of variability in radiomics study design that hinder the comparison of the different model performance and, therefore, clinical implementation. Subsequently, we conducted a use-case meta-analysis using homogenous studies to assess the overall performance of radiomics in estimating programmed death-ligand 1 (PD-L1) expression. Our findings indicate that, despite numerous attempts to predict immunotherapy response, only a limited number of studies share comparable methodologies and report sufficient data about cohorts and methods to be suitable for meta-analysis. Nevertheless, although only a few studies meet these criteria, their promising results underscore the importance of ongoing standardization and benchmarking efforts. This review highlights the importance of uniformity in study design and reporting. Such standardization is crucial to enable meaningful comparisons and demonstrate the validity of biomarkers across diverse populations, facilitating their implementation into the immunotherapy patient selection process.
Collapse
Affiliation(s)
- Marta Ligero
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Bente Gielen
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Victor Navarro
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Pablo Cresta Morgado
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Medical Oncology, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain
- Prostate Cancer Translational Research Group, Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Barcelona, Spain
| | - Olivia Prior
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Rodrigo Dienstmann
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Paolo Nuciforo
- Molecular Oncology Group, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain
| | - Stefano Trebeschi
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Regina Beets-Tan
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Evis Sala
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Scienze Radiologiche ed Ematologiche, Universita Cattolica del Sacro Cuore, Rome, Italy
| | - Elena Garralda
- Department of Medical Oncology, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain
| | - Raquel Perez-Lopez
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| |
Collapse
|
5
|
Li J, Cao Y, Liu Y, Yu L, Zhang Z, Wang X, Bai H, Zhang Y, Liu S, Gao M, Lu C, Li C, Guan Y, Tao Z, Wu Z, Chen J, Yuan Z. Multiomics profiling reveals the benefits of gamma-delta (γδ) T lymphocytes for improving the tumor microenvironment, immunotherapy efficacy and prognosis in cervical cancer. J Immunother Cancer 2024; 12:e008355. [PMID: 38199610 PMCID: PMC10806547 DOI: 10.1136/jitc-2023-008355] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND As an unconventional subpopulation of T lymphocytes, γδ T cells can recognize antigens independently of major histocompatibility complex restrictions. Recent studies have indicated that γδ T cells play contrasting roles in tumor microenvironments-promoting tumor progression in some cancers (eg, gallbladder and leukemia) while suppressing it in others (eg, lung and gastric). γδ T cells are mainly enriched in peripheral mucosal tissues. As the cervix is a mucosa-rich tissue, the role of γδ T cells in cervical cancer warrants further investigation. METHODS We employed a multiomics strategy that integrated abundant data from single-cell and bulk transcriptome sequencing, whole exome sequencing, genotyping array, immunohistochemistry, and MRI. RESULTS Heterogeneity was observed in the level of γδ T-cell infiltration in cervical cancer tissues, mainly associated with the tumor somatic mutational landscape. Definitely, γδ T cells play a beneficial role in the prognosis of patients with cervical cancer. First, γδ T cells exert direct cytotoxic effects in the tumor microenvironment of cervical cancer through the dynamic evolution of cellular states at both poles. Second, higher levels of γδ T-cell infiltration also shape the microenvironment of immune activation with cancer-suppressive properties. We found that these intricate features can be observed by MRI-based radiomics models to non-invasively assess γδ T-cell proportions in tumor tissues in patients. Importantly, patients with high infiltration levels of γδ T cells may be more amenable to immunotherapies including immune checkpoint inhibitors and autologous tumor-infiltrating lymphocyte therapies, than to chemoradiotherapy. CONCLUSIONS γδ T cells play a beneficial role in antitumor immunity in cervical cancer. The abundance of γδ T cells in cervical cancerous tissue is associated with higher response rates to immunotherapy.
Collapse
Affiliation(s)
- Junyi Li
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin, China
| | - Yuanjie Cao
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin, China
| | - Yancheng Liu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin, China
| | - Lu Yu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin, China
| | - Zhen Zhang
- Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Xiaofeng Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin, China
| | - Hui Bai
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin, China
| | - Yuhan Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin, China
| | - Shaochuan Liu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin, China
| | - Miaomiao Gao
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin, China
| | - Chenglu Lu
- Department of Pathology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Chen Li
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin, China
| | - Yong Guan
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin, China
| | - Zhen Tao
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin, China
| | - Zhiqiang Wu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin, China
| | - Jie Chen
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin, China
| | - Zhiyong Yuan
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
6
|
Ghimire P, Kinnersley B, Karami G, Arumugam P, Houlston R, Ashkan K, Modat M, Booth TC. Radiogenomic biomarkers for immunotherapy in glioblastoma: A systematic review of magnetic resonance imaging studies. Neurooncol Adv 2024; 6:vdae055. [PMID: 38680991 PMCID: PMC11046988 DOI: 10.1093/noajnl/vdae055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024] Open
Abstract
Background Immunotherapy is an effective "precision medicine" treatment for several cancers. Imaging signatures of the underlying genome (radiogenomics) in glioblastoma patients may serve as preoperative biomarkers of the tumor-host immune apparatus. Validated biomarkers would have the potential to stratify patients during immunotherapy clinical trials, and if trials are beneficial, facilitate personalized neo-adjuvant treatment. The increased use of whole genome sequencing data, and the advances in bioinformatics and machine learning make such developments plausible. We performed a systematic review to determine the extent of development and validation of immune-related radiogenomic biomarkers for glioblastoma. Methods A systematic review was performed following PRISMA guidelines using the PubMed, Medline, and Embase databases. Qualitative analysis was performed by incorporating the QUADAS 2 tool and CLAIM checklist. PROSPERO registered: CRD42022340968. Extracted data were insufficiently homogenous to perform a meta-analysis. Results Nine studies, all retrospective, were included. Biomarkers extracted from magnetic resonance imaging volumes of interest included apparent diffusion coefficient values, relative cerebral blood volume values, and image-derived features. These biomarkers correlated with genomic markers from tumor cells or immune cells or with patient survival. The majority of studies had a high risk of bias and applicability concerns regarding the index test performed. Conclusions Radiogenomic immune biomarkers have the potential to provide early treatment options to patients with glioblastoma. Targeted immunotherapy, stratified by these biomarkers, has the potential to allow individualized neo-adjuvant precision treatment options in clinical trials. However, there are no prospective studies validating these biomarkers, and interpretation is limited due to study bias with little evidence of generalizability.
Collapse
Affiliation(s)
- Prajwal Ghimire
- Department of Neurosurgery, Kings College Hospital NHS Foundation Trust, London, UK
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| | - Ben Kinnersley
- Department of Oncology, University College London, London, UK
| | | | | | - Richard Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, UK
| | - Keyoumars Ashkan
- Department of Neurosurgery, Kings College Hospital NHS Foundation Trust, London, UK
| | - Marc Modat
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| | - Thomas C Booth
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| |
Collapse
|
7
|
Rossi E, Boldrini L, Maratta MG, Gatta R, Votta C, Tortora G, Schinzari G. Radiomics to predict immunotherapy efficacy in advanced renal cell carcinoma: A retrospective study. Hum Vaccin Immunother 2023; 19:2172926. [PMID: 36723981 PMCID: PMC10012916 DOI: 10.1080/21645515.2023.2172926] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Immunotherapy has become a cornerstone for the treatment of renal cell carcinoma. Nevertheless, some patients are resistant to immune checkpoint inhibitors. The possibility to identify patients who cannot benefit from immunotherapy is a relevant clinical challenge. We analyzed the association between several radiomics features and response to immunotherapy in 53 patients treated with checkpoint inhibitors for advanced renal cell carcinoma. We found that the following features are associated with progression of disease as best tumor response: F_stat.range (p < .0004), F_stat.max (p < .0007), F_stat.var (p < .0016), F_stat.uniformity (p < .0020), F_stat.90thpercentile (p < .0050). Gross tumor volumes characterized by high values of F_stat.var and F_stat.max (greater than 60,000 and greater than 300, respectively) are most likely related to a high risk of progression. Further analyses are warranted to confirm these results. Radiomics, together with other potential predictive factors, such as gut microbiota, genetic features or circulating immune molecules, could allow a personalized treatment for patients with advanced renal cell carcinoma.
Collapse
Affiliation(s)
- Ernesto Rossi
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Luca Boldrini
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Maria Grazia Maratta
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Roberto Gatta
- Dipartimento di Scienze Cliniche e Sperimentali, Universitá degli Studi di Brescia, Brescia, Italy.,Department of Oncology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Claudio Votta
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giampaolo Tortora
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Schinzari
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
8
|
Baxevanis CN. Immune Checkpoint Inhibitors in Cancer Therapy-How Can We Improve Clinical Benefits? Cancers (Basel) 2023; 15:cancers15030881. [PMID: 36765836 PMCID: PMC9913410 DOI: 10.3390/cancers15030881] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are in the spotlight of cancer treatment by increasing the probability for long-term survival in patients with metastatic disease and by considerably prolonging progression-free survival in patients at early disease stages [...].
Collapse
Affiliation(s)
- Constantin N Baxevanis
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
| |
Collapse
|
9
|
Ng J, Gregucci F, Pennell RT, Nagar H, Golden EB, Knisely JPS, Sanfilippo NJ, Formenti SC. MRI-LINAC: A transformative technology in radiation oncology. Front Oncol 2023; 13:1117874. [PMID: 36776309 PMCID: PMC9911688 DOI: 10.3389/fonc.2023.1117874] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/16/2023] [Indexed: 01/28/2023] Open
Abstract
Advances in radiotherapy technologies have enabled more precise target guidance, improved treatment verification, and greater control and versatility in radiation delivery. Amongst the recent novel technologies, Magnetic Resonance Imaging (MRI) guided radiotherapy (MRgRT) may hold the greatest potential to improve the therapeutic gains of image-guided delivery of radiation dose. The ability of the MRI linear accelerator (LINAC) to image tumors and organs with on-table MRI, to manage organ motion and dose delivery in real-time, and to adapt the radiotherapy plan on the day of treatment while the patient is on the table are major advances relative to current conventional radiation treatments. These advanced techniques demand efficient coordination and communication between members of the treatment team. MRgRT could fundamentally transform the radiotherapy delivery process within radiation oncology centers through the reorganization of the patient and treatment team workflow process. However, the MRgRT technology currently is limited by accessibility due to the cost of capital investment and the time and personnel allocation needed for each fractional treatment and the unclear clinical benefit compared to conventional radiotherapy platforms. As the technology evolves and becomes more widely available, we present the case that MRgRT has the potential to become a widely utilized treatment platform and transform the radiation oncology treatment process just as earlier disruptive radiation therapy technologies have done.
Collapse
Affiliation(s)
- John Ng
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States,*Correspondence: John Ng,
| | - Fabiana Gregucci
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States,Department of Radiation Oncology, Miulli General Regional Hospital, Acquaviva delle Fonti, Bari, Italy
| | - Ryan T. Pennell
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States
| | - Himanshu Nagar
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States
| | - Encouse B. Golden
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States
| | | | | | - Silvia C. Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|