1
|
Bergeron P, Milliat F, Deutsch E, Mondini M. Heterogeneous intratumor irradiation: a new partner for immunotherapy. Oncoimmunology 2024; 13:2434280. [PMID: 39589158 PMCID: PMC11601051 DOI: 10.1080/2162402x.2024.2434280] [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: 11/17/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024] Open
Abstract
We recently demonstrated that a heterogeneous tumor irradiation strategy, combining high-dose and low-dose radiotherapy (RT) within the same tumor volume, can synergize with immunotherapy in mice. Our findings indicate that heterogeneous RT doses may promote the spatial diversification of the antitumor immune response. Spatial fractionation of the RT dose has the potential to enhance the therapeutic index of RT/IO combinations, particularly in scenarios where irradiating the entire tumor volume is unfeasible or excessively harmful to the patient.
Collapse
Affiliation(s)
- Paul Bergeron
- INSERM U1030, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Fabien Milliat
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PSE-SANTE/SERAMED/LRMed, Fontenay-aux-Roses, France
| | - Eric Deutsch
- INSERM U1030, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Michele Mondini
- INSERM U1030, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
2
|
Mavrikios A, Baldini C, Loriot Y, Hénon C, Marabelle A, Postel-Vinay S, Champiat S, Danlos FX, Quevrin C, Lopes E, Gazzah A, Bahleda R, Massard C, Deutsch E, Levy A. Is Local Ablative Stereotactic Radiation Therapy a Valuable Rescue Strategy for Time on Drug in Patients Enrolled in Phase I Trials? Int J Radiat Oncol Biol Phys 2024; 120:1245-1256. [PMID: 39128580 DOI: 10.1016/j.ijrobp.2024.07.2336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/13/2024]
Abstract
PURPOSE Patients with advanced tumors enrolled in phase I trials display strong treatment expectations and few therapeutic alternatives. When oligoacquired resistance (OAR; ≤3 lesions of disease progression) occurs, local ablative stereotactic radiation therapy (SRT) could allow disease control and continuing the experimental systemic treatment. METHODS AND MATERIALS Data from patients enrolled in phase I trials evaluating systemic treatments, who experienced OAR while on the phase I systemic therapy and subsequently received SRT between January 2014 and April 2023, were retrospectively analyzed. Progression-free survival (PFS)1 (trial entry to OAR), PFS2 (SRT to first subsequent relapse), time to next systemic treatment (TTNT), and overall survival (OS) were assessed. First subsequent patterns of relapse after SRT were distinguished as OAR2, which could be locally rechallenged, or systemic acquired resistance (SAR; >3 lesions of disease progression). When available, correlations between molecular profile and pathway enrichments of OAR and SAR were explored. RESULTS Forty-two patients with 52 oligoprogressive lesions were analyzed. The median follow-up was 24 months. SRT allowed a median PFS2 of 7.1 months and a median TTNT of 12.8 months. PFS2 included 49% OAR2 and 51% SAR. Median time to first subsequent relapse (9.6 vs 3.5 months; P = .014) and TTNT (22.4 vs 7.6 months; P < .001) were longer for OAR2 compared with that for SAR. No severe toxicities were reported. A PFS1 of <6 months and de novo oligoprogressive lesions were associated with the presence of SAR. More diverse enriched gene pathways were observed for SAR compared with that for OAR2. CONCLUSIONS In patients enrolled in phase I trials, OAR managed with SRT may increase time on investigational systemic treatments. Predictive factors reflecting tumor aggressiveness and clonal heterogeneity could help deciphering OAR2 from SAR and maximize SRT output in the oligoprogressive setting.
Collapse
Affiliation(s)
- Antoine Mavrikios
- Department of Radiation Oncology, International Center for Thoracic Cancers (CICT), Gustave Roussy, Villejuif, France; Sorbonne Université, Faculté de Médecine, Paris, France
| | - Capucine Baldini
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | - Yohann Loriot
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Kremlin-Bicêtre, France
| | - Clémence Hénon
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | - Aurélien Marabelle
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Kremlin-Bicêtre, France
| | - Sophie Postel-Vinay
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Kremlin-Bicêtre, France; Université Paris-Saclay, INSERM U981, Molecular predictors and new targets in oncology, Gustave Roussy, Villejuif, France; University College of London Cancer Institute, London, England
| | - Stéphane Champiat
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | | | - Clément Quevrin
- Université Paris-Saclay, INSERM U1030, Molecular radiotherapy and therapeutic innovation, Gustave Roussy, Villejuif, France
| | - Eloise Lopes
- Université Paris-Saclay, INSERM U1030, Molecular radiotherapy and therapeutic innovation, Gustave Roussy, Villejuif, France
| | - Anas Gazzah
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | - Rastislav Bahleda
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | - Christophe Massard
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Kremlin-Bicêtre, France; Université Paris-Saclay, INSERM U1030, Molecular radiotherapy and therapeutic innovation, Gustave Roussy, Villejuif, France
| | - Eric Deutsch
- Department of Radiation Oncology, International Center for Thoracic Cancers (CICT), Gustave Roussy, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Kremlin-Bicêtre, France; Université Paris-Saclay, INSERM U1030, Molecular radiotherapy and therapeutic innovation, Gustave Roussy, Villejuif, France
| | - Antonin Levy
- Department of Radiation Oncology, International Center for Thoracic Cancers (CICT), Gustave Roussy, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Kremlin-Bicêtre, France; Université Paris-Saclay, INSERM U1030, Molecular radiotherapy and therapeutic innovation, Gustave Roussy, Villejuif, France.
| |
Collapse
|
3
|
Guo Y, Li T, Gong B, Hu Y, Wang S, Yang L, Zheng C. From Images to Genes: Radiogenomics Based on Artificial Intelligence to Achieve Non-Invasive Precision Medicine in Cancer Patients. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2408069. [PMID: 39535476 DOI: 10.1002/advs.202408069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/19/2024] [Indexed: 11/16/2024]
Abstract
With the increasing demand for precision medicine in cancer patients, radiogenomics emerges as a promising frontier. Radiogenomics is originally defined as a methodology for associating gene expression information from high-throughput technologies with imaging phenotypes. However, with advancements in medical imaging, high-throughput omics technologies, and artificial intelligence, both the concept and application of radiogenomics have significantly broadened. In this review, the history of radiogenomics is enumerated, related omics technologies, the five basic workflows and their applications across tumors, the role of AI in radiogenomics, the opportunities and challenges from tumor heterogeneity, and the applications of radiogenomics in tumor immune microenvironment. The application of radiogenomics in positron emission tomography and the role of radiogenomics in multi-omics studies is also discussed. Finally, the challenges faced by clinical transformation, along with future trends in this field is discussed.
Collapse
Affiliation(s)
- Yusheng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Tianxiang Li
- Department of Ultrasound, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical. Sciences, Peking Union Medical College, Beijing, 100730, China
| | - Bingxin Gong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yan Hu
- Research Institute of Trustworthy Autonomous Systems and Department of Computer Science and Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Sichen Wang
- School of Life Science and Technology, Computational Biology Research Center, Harbin Institute of Technology, Harbin, 150001, China
| | - Lian Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| |
Collapse
|
4
|
Deutsch E, Levy A. Eradicating gross tumor disease: a prerequisite for efficient radioimmunotherapy? J Natl Cancer Inst 2024; 116:1008-1011. [PMID: 38539049 DOI: 10.1093/jnci/djae071] [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: 09/26/2023] [Revised: 11/22/2023] [Accepted: 03/18/2024] [Indexed: 07/06/2024] Open
Abstract
Radiation therapy may induce off-target antitumor "abscopal" immunostimulatory and immunosuppressive effects. Several preclinical and early clinical studies revealed promising results when combining radiation therapy with immunostimulatory agents. Most radioimmunotherapy randomized trials showed disappointing results in patients with advanced tumors. In contrast, outcomes were encouraging when immunotherapy was delivered on top of gross disease elimination with curative-intent radiation therapy. In this review, we highlight available results from randomized trials and discuss the potential impact of overall tumor burden on the observed efficacy of radioimmunotherapy.
Collapse
Affiliation(s)
- Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, INSERM U1030, Molecular Radiotherapy and Therapeutic Innovation, Villejuif, France
- Université Paris-Saclay, Faculté de Médecine, Kremlin-Bicêtre, France
| | - Antonin Levy
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, INSERM U1030, Molecular Radiotherapy and Therapeutic Innovation, Villejuif, France
- Université Paris-Saclay, Faculté de Médecine, Kremlin-Bicêtre, France
| |
Collapse
|
5
|
Hijazi A, Galon J. Principles of risk assessment in colon cancer: immunity is key. Oncoimmunology 2024; 13:2347441. [PMID: 38694625 PMCID: PMC11062361 DOI: 10.1080/2162402x.2024.2347441] [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: 11/30/2023] [Accepted: 04/16/2024] [Indexed: 05/04/2024] Open
Abstract
In clinical practice, the administration of adjuvant chemotherapy (ACT) following tumor surgical resection raises a critical dilemma for stage II colon cancer (CC) patients. The prognostic features used to identify high-risk CC patients rely on the pathological assessment of tumor cells. Currently, these factors are considered for stratifying patients who may benefit from ACT at early CC stages. However, the extent to which these factors predict clinical outcomes (i.e. recurrence, survival) remains highly controversial, also uncertainty persists regarding patients' response to treatment, necessitating further investigation. Therefore, an imperious need is to explore novel biomarkers that can reliably stratify patients at risk, to optimize adjuvant treatment decisions. Recently, we evaluated the prognostic and predictive value of Immunoscore (IS), an immune digital-pathology assay, in stage II CC patients. IS emerged as the sole significant parameter for predicting disease-free survival (DFS) in high-risk patients. Moreover, IS effectively stratified patients who would benefit most from ACT based on their risk of recurrence, thus predicting their outcomes. Notably, our findings revealed that digital IS outperformed the visual quantitative assessment of the immune response conducted by expert pathologists. The latest edition of the WHO classification for digestive tumor has introduced the evaluation of the immune response, as assessed by IS, as desirable and essential diagnostic criterion. This supports the revision of current cancer guidelines and strongly recommends the implementation of IS into clinical practice as a patient stratification tool, to guide CC treatment decisions. This approach may provide appropriate personalized therapeutic decisions that could critically impact early-stage CC patient care.
Collapse
Affiliation(s)
- Assia Hijazi
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris, France
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris, France
- Veracyte, Marseille, France
| |
Collapse
|
6
|
Tompkins A, Gray ZN, Dadey RE, Zenkin S, Batavani N, Newman S, Amouzegar A, Ak M, Ak N, Pak TY, Peddagangireddy V, Mamindla P, Behr S, Goodman A, Ploucha DL, Kirkwood JM, Zarour HM, Najjar YG, Davar D, Colen R, Luke JJ, Bao R. Radiomic analysis of patient and inter-organ heterogeneity in response to immunotherapies and BRAF targeted therapy in metastatic melanoma. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.26.24306411. [PMID: 38712112 PMCID: PMC11071587 DOI: 10.1101/2024.04.26.24306411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Background Variability in treatment response may be attributable to organ-level heterogeneity in tumor lesions. Radiomic analysis of medical images can elucidate non-invasive biomarkers of clinical outcome. Organ-specific radiomic comparison across immunotherapies and targeted therapies has not been previously reported. Methods We queried UPMC Hillman Cancer Center registry for patients with metastatic melanoma (MEL) treated with immune checkpoint inhibitors (ICI) (anti-PD1/CTLA4 [ipilimumab+nivolumab; I+N] or anti-PD1 monotherapy) or BRAF targeted therapy. Best overall response was measured using RECIST v1.1. Lesions were segmented into discrete volume-of-interest with 400 radiomics features extracted. Overall and organ-specific machine-learning models were constructed to predict disease control (DC) versus progressive disease (PD) using XGBoost. Results 291 MEL patients were identified, including 242 ICI (91 I+N, 151 PD1) and 49 BRAF. 667 metastases were analyzed, including 541 ICI (236 I+N, 305 PD1) and 126 BRAF. Across cohorts, baseline demographics included 39-47% female, 24-29% M1C, 24-46% M1D, and 61-80% with elevated LDH. Among patients experiencing DC, the organs with the greatest reduction were liver (-88%±12%, I+N; mean±S.E.M.) and lung (-72%±8%, I+N). For patients with multiple same-organ target lesions, the highest inter-lesion heterogeneity was observed in brain among patients who received ICI while no intra-organ heterogeneity was observed in BRAF. 267 patients were kept for radiomic modeling, including 221 ICI (86 I+N, 135 PD1) and 46 BRAF. Models consisting of optimized radiomic signatures classified DC/PD across I+N (AUC=0.85) and PD1 (0.71) and within individual organ sites (AUC=0.72∼0.94). Integration of clinical variables improved the models' performance. Comparison of models between treatments and across organ sites suggested mostly non-overlapping DC or PD features. Skewness, kurtosis, and informational measure of correlation (IMC) were among the radiomic features shared between overall response models. Kurtosis and IMC were also utilized by multiple organ-site models. Conclusions Differential organ-specific response was observed across BRAF and ICI with within organ heterogeneity observed for ICI but not for BRAF. Radiomic features of organ-specific response demonstrated little overlap. Integrating clinical factors with radiomics improves the prediction of disease course outcome and prediction of tumor heterogeneity.
Collapse
|
7
|
Cui FB, Lv X, Yan CL, Eng WS, Yu SY, Zheng QH. Development and application of a fully automatic multi-function cassette module Mortenon M1 for radiopharmaceutical synthesis. Ann Nucl Med 2024; 38:247-263. [PMID: 38145430 DOI: 10.1007/s12149-023-01893-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 12/05/2023] [Indexed: 12/26/2023]
Abstract
INTRODUCTION Functions of existing automatic module systems for synthesis of radiopharmaceuticals mainly focus on the radiolabeling of small molecules. There are few modules which have achieved full-automatic radiolabeling of non-metallic and metallic nuclides on small molecules, peptides, and antibody drugs. This study aimed to develop and test a full-automatic multifunctional module system for the safe, stable, and efficient production of radiopharmaceuticals. METHODS According to characteristics of labeling process of radioactive drugs, using UG and Solidworks softwares, full-automatic cassette-based synthesis module system Mortenon M1 for synthesis of radiopharmaceuticals with various radionuclides, was designed and tested. Mortenon M1 has at least three significant highlights: the cassettes are disposable, and there is no need of manual cleaning; the synthesis method program is flexible and can be edited freely by users according to special needs; this module system is suitable for radiolabeling of both small-molecule and macromolecular drugs, with potentially various radionuclides including 18F, 64Cu, 68Ga, 89Zr, 177Lu, etc. By program control methods for certain drugs, Mortenon M1 was used for radiolabeling of both small-molecule drugs such as [68Ga]-FAPI-46 and macromolecular drugs such as [89Zr]-TROP2 antibody. Quality control assays for product purity were performed with radio-iTLC and radio-HPLC, and the radiotracers were confirmed for application in microPET imaging in xenograft tumor-bearing mouse models. RESULTS Functional tests for Mortenon M1 module system were conducted, with [68Ga]-FAPI-46 and [89Zr]-TROP2 antibody as goal synthetic products, and it displayed that with the cassette modules, the preset goals could be achieved successfully. The radiolabeling synthesis yield was good ([68Ga]-FAPI-46, 70.63% ± 2.85%, n = 10; [89Zr]-TROP2, 82.31% ± 3.92%, n = 10), and the radiochemical purity via radio-iTLC assay of the radiolabeled products was above 99% after purification. MicroPET imaging results showed that the radiolabeled tracers had reasonable radioactive distribution in MDA-MB-231 and SNU-620 xenograft tumor-bearing mice, and the tumor targeted radiouptake was satisfactory for diagnosis. CONCLUSION This study demonstrated that the full-automatic module system Mortenon M1 is efficient for radiolabeling synthesis of both small-molecule and macromolecular substrates. It may be helpful to reduce radiation exposure for safety, provide qualified radiolabeled products and reliable PET diagnosis, and ensure stable production and supply of radiopharmaceuticals.
Collapse
Affiliation(s)
- Fang-Bo Cui
- Department of Oncology, The People's Hospital of Ma Anshan, Ma Anshan, 243000, Anhui, People's Republic of China
| | - Xuan Lv
- Norroy Bioscience Co., Ltd, Building 2, Lihu Business Park, Zhongbang MOHO, Huize Road, Binhu District, Wuxi, 214000, Jiangsu, People's Republic of China
| | - Cheng-Long Yan
- Norroy Bioscience Co., Ltd, Building 2, Lihu Business Park, Zhongbang MOHO, Huize Road, Binhu District, Wuxi, 214000, Jiangsu, People's Republic of China
| | - Wai-Si Eng
- Norroy Bioscience Co., Ltd, Building 2, Lihu Business Park, Zhongbang MOHO, Huize Road, Binhu District, Wuxi, 214000, Jiangsu, People's Republic of China
| | - Shan-You Yu
- Norroy Bioscience Co., Ltd, Building 2, Lihu Business Park, Zhongbang MOHO, Huize Road, Binhu District, Wuxi, 214000, Jiangsu, People's Republic of China.
| | - Qi-Huang Zheng
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 1345 West 16th Street, Room 112, Indianapolis, IN, 46202, USA.
| |
Collapse
|
8
|
Levy A, Morel D, Texier M, Sun R, Durand-Labrunie J, Rodriguez-Ruiz ME, Racadot S, Supiot S, Magné N, Cyrille S, Louvel G, Massard C, Verlingue L, Bouquet F, Bustillos A, Bouarroudj L, Quevrin C, Clémenson C, Mondini M, Meziani L, Tselikas L, Bahleda R, Hollebecque A, Deutsch E. An international phase II trial and immune profiling of SBRT and atezolizumab in advanced pretreated colorectal cancer. Mol Cancer 2024; 23:61. [PMID: 38519913 PMCID: PMC10960440 DOI: 10.1186/s12943-024-01970-8] [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: 09/19/2023] [Accepted: 02/22/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Immuno-radiotherapy may improve outcomes for patients with advanced solid tumors, although optimized combination modalities remain unclear. Here, we report the colorectal (CRC) cohort analysis from the SABR-PDL1 trial that evaluated the PD-L1 inhibitor atezolizumab in combination with stereotactic body radiation therapy (SBRT) in advanced cancer patients. METHODS Eligible patients received atezolizumab 1200 mg every 3 weeks until progression or unmanageable toxicity, together with ablative SBRT delivered concurrently with the 2nd cycle (recommended dose of 45 Gy in 3 fractions, adapted upon normal tissue tolerance constraint). SBRT was delivered to at least one tumor site, with at least one additional measurable lesion being kept from the radiation field. The primary efficacy endpoint was one-year progression-free survival (PFS) rate from the start of atezolizumab. Sequential tumor biopsies were collected for deep multi-feature immune profiling. RESULTS Sixty pretreated (median of 2 prior lines) advanced CRC patients (38 men [63%]; median age, 59 years [range, 20-81 years]; 77% with liver metastases) were enrolled in five centers (France: n = 4, Spain: n = 1) from 11/2016 to 04/2019. All but one (98%) received atezolizumab and 54/60 (90%) received SBRT. The most frequently irradiated site was lung (n = 30/54; 56.3%). Treatment-related G3 (no G4-5) toxicity was observed in 3 (5%) patients. Median OS and PFS were respectively 8.4 [95%CI:5.9-11.6] and 1.4 months [95%CI:1.2-2.6], including five (9%) patients with PFS > 1 year (median time to progression: 19.2 months, including 2/5 MMR-proficient). Best overall responses consisted of stable disease (n = 38; 64%), partial (n = 3; 5%) and complete response (n = 1; 2%). Immune-centric multiplex IHC and RNAseq showed that SBRT redirected immune cells towards tumor lesions, even in the case of radio-induced lymphopenia. Baseline tumor PD-L1 and IRF1 nuclear expression (both in CD3 + T cells and in CD68 + cells) were higher in responding patients. Upregulation of genes that encode for proteins known to increase T and B cell trafficking to tumors (CCL19, CXCL9), migration (MACF1) and tumor cell killing (GZMB) correlated with responses. CONCLUSIONS This study provides new data on the feasibility, efficacy, and immune context of tumors that may help identifying advanced CRC patients most likely to respond to immuno-radiotherapy. TRIAL REGISTRATION EudraCT N°: 2015-005464-42; Clinicaltrial.gov number: NCT02992912.
Collapse
Affiliation(s)
- Antonin Levy
- Department of Radiation Oncology, Gustave Roussy, 114 Rue E. Vaillant, 94850, Villejuif, France.
- INSERM U1030, Radiothérapie Moléculaire, Villejuif, France.
- Faculty of Medicine, Université Paris Saclay, Le Kremlin-Bicêtre, France.
| | - Daphné Morel
- Department of Radiation Oncology, Gustave Roussy, 114 Rue E. Vaillant, 94850, Villejuif, France
- INSERM U1030, Radiothérapie Moléculaire, Villejuif, France
| | - Matthieu Texier
- Biostatistics and Epidemiology Office, Gustave Roussy, Villejuif, France
- Oncostat 1018 INSERM, University Paris-Saclay, Villejuif, France
| | - Roger Sun
- Department of Radiation Oncology, Gustave Roussy, 114 Rue E. Vaillant, 94850, Villejuif, France
- INSERM U1030, Radiothérapie Moléculaire, Villejuif, France
- Faculty of Medicine, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Jerome Durand-Labrunie
- Department of Radiation Oncology, Gustave Roussy, 114 Rue E. Vaillant, 94850, Villejuif, France
| | | | - Severine Racadot
- Department of Radiation Oncology, Centre Léon Bérard, Lyon, France
| | - Stéphane Supiot
- Department of Radiation Oncology, Institut de Cancérologie de L'Ouest-Centre Rene Gauducheau, St Herblain, Nantes, France
| | - Nicolas Magné
- Department of Radiation Oncology, Institut Bergonié, Bordeaux, France
| | - Stacy Cyrille
- Biostatistics and Epidemiology Office, Gustave Roussy, Villejuif, France
- Oncostat 1018 INSERM, University Paris-Saclay, Villejuif, France
| | - Guillaume Louvel
- Department of Radiation Oncology, Gustave Roussy, 114 Rue E. Vaillant, 94850, Villejuif, France
| | - Christophe Massard
- INSERM U1030, Radiothérapie Moléculaire, Villejuif, France
- Faculty of Medicine, Université Paris Saclay, Le Kremlin-Bicêtre, France
- Drug Development Department (DITEP), Gustave Roussy-Cancer Campus, Villejuif, France
| | - Loic Verlingue
- Drug Development Department (DITEP), Gustave Roussy-Cancer Campus, Villejuif, France
| | - Fanny Bouquet
- Product Development Medical Affairs, F Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Alberto Bustillos
- Product Development Medical Affairs, F Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Lisa Bouarroudj
- Department of Radiation Oncology, Gustave Roussy, 114 Rue E. Vaillant, 94850, Villejuif, France
- INSERM U1030, Radiothérapie Moléculaire, Villejuif, France
- Bioinformatic Platform, Gustave Roussy, Villejuif, France
| | | | | | | | - Lydia Meziani
- INSERM U1030, Radiothérapie Moléculaire, Villejuif, France
| | - Lambros Tselikas
- Faculty of Medicine, Université Paris Saclay, Le Kremlin-Bicêtre, France
- Department of Interventional Radiology, Gustave Roussy, Villejuif, France
| | - Rastilav Bahleda
- Drug Development Department (DITEP), Gustave Roussy-Cancer Campus, Villejuif, France
| | - Antoine Hollebecque
- Drug Development Department (DITEP), Gustave Roussy-Cancer Campus, Villejuif, France
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy, 114 Rue E. Vaillant, 94850, Villejuif, France.
- INSERM U1030, Radiothérapie Moléculaire, Villejuif, France.
- Faculty of Medicine, Université Paris Saclay, Le Kremlin-Bicêtre, France.
| |
Collapse
|
9
|
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
|
10
|
Huang W, Xiong W, Tang L, Chen C, Yuan Q, Zhang C, Zhou K, Sun Z, Zhang T, Han Z, Feng H, Liang X, Zhong Y, Deng H, Yu L, Xu Y, Wang W, Shen L, Li G, Jiang Y. Non-invasive CT imaging biomarker to predict immunotherapy response in gastric cancer: a multicenter study. J Immunother Cancer 2023; 11:e007807. [PMID: 38179695 PMCID: PMC10668251 DOI: 10.1136/jitc-2023-007807] [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] [Accepted: 10/24/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Despite remarkable benefits have been provided by immune checkpoint inhibitors in gastric cancer (GC), predictions of treatment response and prognosis remain unsatisfactory, making identifying biomarkers desirable. The aim of this study was to develop and validate a CT imaging biomarker to predict the immunotherapy response in patients with GC and investigate the associated immune infiltration patterns. METHODS This retrospective study included 294 GC patients who received anti-PD-1/PD-L1 immunotherapy from three independent medical centers between January 2017 and April 2022. A radiomics score (RS) was developed from the intratumoral and peritumoral features on pretreatment CT images to predict immunotherapy-related progression-free survival (irPFS). The performance of the RS was evaluated by the area under the time-dependent receiver operating characteristic curve (AUC). Multivariable Cox regression analysis was performed to construct predictive nomogram of irPFS. The C-index was used to determine the performance of the nomogram. Bulk RNA sequencing of tumors from 42 patients in The Cancer Genome Atlas was used to investigate the RS-associated immune infiltration patterns. RESULTS Overall, 89 of 294 patients (median age, 57 years (IQR 48-66 years); 171 males) had an objective response to immunotherapy. The RS included 13 CT features that yielded AUCs of 12-month irPFS of 0.787, 0.810 and 0.785 in the training, internal validation, and external validation 1 cohorts, respectively, and an AUC of 24-month irPFS of 0.805 in the external validation 2 cohort. Patients with low RS had longer irPFS in each cohort (p<0.05). Multivariable Cox regression analyses showed RS is an independent prognostic factor of irPFS. The nomogram that integrated the RS and clinical characteristics showed improved performance in predicting irPFS, with C-index of 0.687-0.778 in the training and validation cohorts. The CT imaging biomarker was associated with M1 macrophage infiltration. CONCLUSION The findings of this prognostic study suggest that the non-invasive CT imaging biomarker can effectively predict immunotherapy outcomes in patients with GC and is associated with innate immune signaling, which can serve as a potential tool for individual treatment decisions.
Collapse
Affiliation(s)
- Weicai Huang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Wenjun Xiong
- Department of Gastrointestinal Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lei Tang
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Chuanli Chen
- Department of Medical Imaging Center, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Qingyu Yuan
- Department of Medical Imaging Center, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Cheng Zhang
- Department of Gastrointestinal Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Kangneng Zhou
- University of Science and Technology, Beijing, China
| | - Zepang Sun
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Taojun Zhang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Zhen Han
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Hao Feng
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Xiaokun Liang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- Shenzhen Colleges of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Yonghong Zhong
- Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haijun Deng
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Lequan Yu
- Department of Statistics and Actuarial Science, The University of Hong Kong, Hong Kong, China
| | - Yikai Xu
- Department of Medical Imaging Center, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Wei Wang
- Department of Gastrointestinal Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Guoxin Li
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Yuming Jiang
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
11
|
Nguyen TM, Bertolus C, Giraud P, Burgun A, Saintigny P, Bibault JE, Foy JP. A Radiomics Approach to Identify Immunologically Active Tumor in Patients with Head and Neck Squamous Cell Carcinomas. Cancers (Basel) 2023; 15:5369. [PMID: 38001629 PMCID: PMC10670096 DOI: 10.3390/cancers15225369] [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: 09/21/2023] [Revised: 11/05/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND We recently developed a gene-expression-based HOT score to identify the hot/cold phenotype of head and neck squamous cell carcinomas (HNSCCs), which is associated with the response to immunotherapy. Our goal was to determine whether radiomic profiling from computed tomography (CT) scans can distinguish hot and cold HNSCC. METHOD We included 113 patients from The Cancer Genome Atlas (TCGA) and 20 patients from the Groupe Hospitalier Pitié-Salpêtrière (GHPS) with HNSCC, all with available pre-treatment CT scans. The hot/cold phenotype was computed for all patients using the HOT score. The IBEX software (version 4.11.9, accessed on 30 march 2020) was used to extract radiomic features from the delineated tumor region in both datasets, and the intraclass correlation coefficient (ICC) was computed to select robust features. Machine learning classifier models were trained and tested in the TCGA dataset and validated using the area under the receiver operator characteristic curve (AUC) in the GHPS cohort. RESULTS A total of 144 radiomic features with an ICC >0.9 was selected. An XGBoost model including these selected features showed the best performance prediction of the hot/cold phenotype with AUC = 0.86 in the GHPS validation dataset. CONCLUSIONS AND RELEVANCE We identified a relevant radiomic model to capture the overall hot/cold phenotype of HNSCC. This non-invasive approach could help with the identification of patients with HNSCC who may benefit from immunotherapy.
Collapse
Affiliation(s)
- Tan Mai Nguyen
- Sorbonne Université, Department of Maxillo-Facial Surgery, Hôpital Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, 75013 Paris, France; (T.M.N.); (C.B.)
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France;
- INSERM, UMR S1138, Cordeliers Research Center, Université Paris Cité, 75005 Paris, France; (P.G.); (A.B.); (J.-E.B.)
| | - Chloé Bertolus
- Sorbonne Université, Department of Maxillo-Facial Surgery, Hôpital Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, 75013 Paris, France; (T.M.N.); (C.B.)
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France;
| | - Paul Giraud
- INSERM, UMR S1138, Cordeliers Research Center, Université Paris Cité, 75005 Paris, France; (P.G.); (A.B.); (J.-E.B.)
- Sorbonne Université, Department of Radiation Oncology, Hôpital Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, 75013 Paris, France
| | - Anita Burgun
- INSERM, UMR S1138, Cordeliers Research Center, Université Paris Cité, 75005 Paris, France; (P.G.); (A.B.); (J.-E.B.)
| | - Pierre Saintigny
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France;
- Department of Medical Oncology, Centre Léon Bérard, 69008 Lyon, France
| | - Jean-Emmanuel Bibault
- INSERM, UMR S1138, Cordeliers Research Center, Université Paris Cité, 75005 Paris, France; (P.G.); (A.B.); (J.-E.B.)
- Department of Radiation Oncology, Hôpital Européen Georges-Pompidou, Université Paris Cité, 75015 Paris, France
| | - Jean-Philippe Foy
- Sorbonne Université, Department of Maxillo-Facial Surgery, Hôpital Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, 75013 Paris, France; (T.M.N.); (C.B.)
- Sorbonne Université, INSERM UMRS 938, Centre de Recherche de Saint Antoine, Team Cancer Biology and Therapeutics, 75011 Paris, France
| |
Collapse
|
12
|
McGale J, Hama J, Yeh R, Vercellino L, Sun R, Lopci E, Ammari S, Dercle L. Artificial Intelligence and Radiomics: Clinical Applications for Patients with Advanced Melanoma Treated with Immunotherapy. Diagnostics (Basel) 2023; 13:3065. [PMID: 37835808 PMCID: PMC10573034 DOI: 10.3390/diagnostics13193065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 10/15/2023] Open
Abstract
Immunotherapy has greatly improved the outcomes of patients with metastatic melanoma. However, it has also led to new patterns of response and progression, creating an unmet need for better biomarkers to identify patients likely to achieve a lasting clinical benefit or experience immune-related adverse events. In this study, we performed a focused literature survey covering the application of artificial intelligence (AI; in the form of radiomics, machine learning, and deep learning) to patients diagnosed with melanoma and treated with immunotherapy, reviewing 12 studies relevant to the topic published up to early 2022. The most commonly investigated imaging modality was CT imaging in isolation (n = 9, 75.0%), while patient cohorts were most frequently recruited retrospectively and from single institutions (n = 7, 58.3%). Most studies concerned the development of AI tools to assist in prognostication (n = 5, 41.7%) or the prediction of treatment response (n = 6, 50.0%). Validation methods were disparate, with two studies (16.7%) performing no validation and equal numbers using cross-validation (n = 3, 25%), a validation set (n = 3, 25%), or a test set (n = 3, 25%). Only one study used both validation and test sets (n = 1, 8.3%). Overall, promising results have been observed for the application of AI to immunotherapy-treated melanoma. Further improvement and eventual integration into clinical practice may be achieved through the implementation of rigorous validation using heterogeneous, prospective patient cohorts.
Collapse
Affiliation(s)
- Jeremy McGale
- Department of Radiology, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Jakob Hama
- Queens Hospital Center, Icahn School of Medicine at Mt. Sinai, Queens, NY 10029, USA
| | - Randy Yeh
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Laetitia Vercellino
- Nuclear Medicine Department, INSERM UMR S942, Hôpital Saint-Louis, Assistance-Publique, Hôpitaux de Paris, Université Paris Cité, 75010 Paris, France
| | - Roger Sun
- Department of Radiation Oncology, Gustave Roussy, 94800 Villejuif, France
| | - Egesta Lopci
- Nuclear Medicine Unit, IRCCS—Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Samy Ammari
- Department of Medical Imaging, BIOMAPS, UMR1281 INSERM, CEA, CNRS, Gustave Roussy, Université Paris-Saclay, 94800 Villejuif, France
- ELSAN Department of Radiology, Institut de Cancérologie Paris Nord, 95200 Sarcelles, France
| | - Laurent Dercle
- Department of Radiology, New York-Presbyterian Hospital, New York, NY 10032, USA
| |
Collapse
|
13
|
Liu Z, Duan T, Zhang Y, Weng S, Xu H, Ren Y, Zhang Z, Han X. Radiogenomics: a key component of precision cancer medicine. Br J Cancer 2023; 129:741-753. [PMID: 37414827 PMCID: PMC10449908 DOI: 10.1038/s41416-023-02317-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 05/02/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023] Open
Abstract
Radiogenomics, focusing on the relationship between genomics and imaging phenotypes, has been widely applied to address tumour heterogeneity and predict immune responsiveness and progression. It is an inevitable consequence of current trends in precision medicine, as radiogenomics costs less than traditional genetic sequencing and provides access to whole-tumour information rather than limited biopsy specimens. By providing voxel-by-voxel genetic information, radiogenomics can allow tailored therapy targeting a complete, heterogeneous tumour or set of tumours. In addition to quantifying lesion characteristics, radiogenomics can also be used to distinguish benign from malignant entities, as well as patient characteristics, to better stratify patients according to disease risk, thereby enabling more precise imaging and screening. Here, we have characterised the radiogenomic application in precision medicine using a multi-omic approach. we outline the main applications of radiogenomics in diagnosis, treatment planning and evaluations in the field of oncology with the aim of developing quantitative and personalised medicine. Finally, we discuss the challenges in the field of radiogenomics and the scope and clinical applicability of these methods.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Interventional Institute of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, 450052, Zhengzhou, Henan, China
| | - Tian Duan
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
- Interventional Institute of Zhengzhou University, 450052, Zhengzhou, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, 450052, Zhengzhou, Henan, China.
| |
Collapse
|
14
|
Tonneau M, Phan K, Manem VSK, Low-Kam C, Dutil F, Kazandjian S, Vanderweyen D, Panasci J, Malo J, Coulombe F, Gagné A, Elkrief A, Belkaïd W, Di Jorio L, Orain M, Bouchard N, Muanza T, Rybicki FJ, Kafi K, Huntsman D, Joubert P, Chandelier F, Routy B. Generalization optimizing machine learning to improve CT scan radiomics and assess immune checkpoint inhibitors' response in non-small cell lung cancer: a multicenter cohort study. Front Oncol 2023; 13:1196414. [PMID: 37546399 PMCID: PMC10400292 DOI: 10.3389/fonc.2023.1196414] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/28/2023] [Indexed: 08/08/2023] Open
Abstract
Background Recent developments in artificial intelligence suggest that radiomics may represent a promising non-invasive biomarker to predict response to immune checkpoint inhibitors (ICIs). Nevertheless, validation of radiomics algorithms in independent cohorts remains a challenge due to variations in image acquisition and reconstruction. Using radiomics, we investigated the importance of scan normalization as part of a broader machine learning framework to enable model external generalizability to predict ICI response in non-small cell lung cancer (NSCLC) patients across different centers. Methods Radiomics features were extracted and compared from 642 advanced NSCLC patients on pre-ICI scans using established open-source PyRadiomics and a proprietary DeepRadiomics deep learning technology. The population was separated into two groups: a discovery cohort of 512 NSCLC patients from three academic centers and a validation cohort that included 130 NSCLC patients from a fourth center. We harmonized images to account for variations in reconstruction kernel, slice thicknesses, and device manufacturers. Multivariable models, evaluated using cross-validation, were used to estimate the predictive value of clinical variables, PD-L1 expression, and PyRadiomics or DeepRadiomics for progression-free survival at 6 months (PFS-6). Results The best prognostic factor for PFS-6, excluding radiomics features, was obtained with the combination of Clinical + PD-L1 expression (AUC = 0.66 in the discovery and 0.62 in the validation cohort). Without image harmonization, combining Clinical + PyRadiomics or DeepRadiomics delivered an AUC = 0.69 and 0.69, respectively, in the discovery cohort, but dropped to 0.57 and 0.52, in the validation cohort. This lack of generalizability was consistent with observations in principal component analysis clustered by CT scan parameters. Subsequently, image harmonization eliminated these clusters. The combination of Clinical + DeepRadiomics reached an AUC = 0.67 and 0.63 in the discovery and validation cohort, respectively. Conversely, the combination of Clinical + PyRadiomics failed generalizability validations, with AUC = 0.66 and 0.59. Conclusion We demonstrated that a risk prediction model combining Clinical + DeepRadiomics was generalizable following CT scan harmonization and machine learning generalization methods. These results had similar performances to routine oncology practice using Clinical + PD-L1. This study supports the strong potential of radiomics as a future non-invasive strategy to predict ICI response in advanced NSCLC.
Collapse
Affiliation(s)
- Marion Tonneau
- Department of Cancer Research, Centre de Recherche du Centre Hospitalier Universitaire de Montréal (CRCHUM), Montreal, QC, Canada
- Université de Médecine, Lille, France
| | - Kim Phan
- Imagia Canexia Health, Montreal, QC, Canada
| | - Venkata S. K. Manem
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Université Laval, Québec City, QC, Canada
- Department of Mathematics and Computer Science, University of Quebec at Trois-Rivières, Trois-Rivières, QC, Canada
| | | | | | - Suzanne Kazandjian
- Department of Medical Oncology, Jewish General Hospital, Montreal, QC, Canada
| | - Davy Vanderweyen
- Department of Radiology, Centre Hospitalier de Sherbrooke (CHUS), Sherbrooke, QC, Canada
| | - Justin Panasci
- Department of Medical Oncology, Jewish General Hospital, Montreal, QC, Canada
| | - Julie Malo
- Department of Cancer Research, Centre de Recherche du Centre Hospitalier Universitaire de Montréal (CRCHUM), Montreal, QC, Canada
| | - François Coulombe
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Université Laval, Québec City, QC, Canada
| | - Andréanne Gagné
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Université Laval, Québec City, QC, Canada
| | - Arielle Elkrief
- Department of Cancer Research, Centre de Recherche du Centre Hospitalier Universitaire de Montréal (CRCHUM), Montreal, QC, Canada
- Hemato-Oncology Division, Centre Hospitalier de l’université de Montreal, Montreal, QC, Canada
| | - Wiam Belkaïd
- Department of Cancer Research, Centre de Recherche du Centre Hospitalier Universitaire de Montréal (CRCHUM), Montreal, QC, Canada
| | | | - Michele Orain
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Université Laval, Québec City, QC, Canada
| | - Nicole Bouchard
- Department of Oncology, Centre Hospitalier de Sherbrooke (CHUS), Sherbrooke, QC, Canada
| | - Thierry Muanza
- Department of Medical Oncology, Jewish General Hospital, Montreal, QC, Canada
- Department of Radiation Oncology, Lady Davis Institute of the Jewish General Hospital, Montreal, QC, Canada
| | | | - Kam Kafi
- Imagia Canexia Health, Montreal, QC, Canada
| | | | - Philippe Joubert
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Université Laval, Québec City, QC, Canada
- Department of Pathology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
| | | | - Bertrand Routy
- Department of Cancer Research, Centre de Recherche du Centre Hospitalier Universitaire de Montréal (CRCHUM), Montreal, QC, Canada
- Hemato-Oncology Division, Centre Hospitalier de l’université de Montreal, Montreal, QC, Canada
| |
Collapse
|
15
|
Zhang Y, Hu HH, Zhou SH, Xia WY, Zhang Y, Zhang JP, Fu XL, Yu W. PET-based radiomics visualizes tumor-infiltrating CD8 T cell exhaustion to optimize radiotherapy/immunotherapy combination in mouse models of lung cancer. Biomark Res 2023; 11:10. [PMID: 36694213 PMCID: PMC9875413 DOI: 10.1186/s40364-023-00454-z] [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: 10/08/2022] [Accepted: 01/13/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Cumulative preclinical and clinical evidences showed radiotherapy might augment systemic antitumoral responses to immunotherapy for metastatic non-small cell lung cancer, but the optimal timing of combination is still unclear. The overall infiltration and exhausted subpopulations of tumor-infiltrating CD8+ T cells might be a potential biomarker indicating the response to immune checkpoint inhibitors (ICI), the alteration of which is previously uncharacterized during peri-irradiation period, while dynamic monitoring is unavailable via repeated biopsies in clinical practice. METHODS Basing on tumor-bearing mice model, we investigated the dynamics of overall infiltration and exhausted subpopulations of CD8+ T cells after ablative irradiation. With the understanding of distinct metabolic characteristics accompanied with T cell exhaustion, we developed a PET radiomics approach to identify and visualize T cell exhaustion status. RESULTS CD8+ T cell infiltration increased from 3 to 14 days after ablative irradiation while terminally exhausted populations significantly predominated CD8+ T cells during late course of this infiltrating period, indicating that 3-7 days post-irradiation might be a potential appropriate window for delivering ICI treatment. A PET radiomics approach was established to differentiate T cell exhaustion status, which fitted well in both ICI and irradiation settings. We also visualized the underlying association of more heterogeneous texture on PET images with progressed T cell exhaustion. CONCLUSIONS We proposed a non-invasive imaging predictor which accurately assessed heterogeneous T cell exhaustion status relevant to ICI treatment and irradiation, and might serve as a promising solution to timely estimate immune-responsiveness of tumor microenvironment and the optimal timing of combined therapy.
Collapse
Affiliation(s)
- Ying Zhang
- grid.412524.40000 0004 0632 3994Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai, 200030 China
| | - Hui-Hui Hu
- grid.412524.40000 0004 0632 3994Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai, 200030 China
| | - Shi-Hong Zhou
- grid.412524.40000 0004 0632 3994Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wu-Yan Xia
- grid.412524.40000 0004 0632 3994Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai, 200030 China
| | - Yan Zhang
- grid.16821.3c0000 0004 0368 8293Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Ping Zhang
- grid.452404.30000 0004 1808 0942Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiao-Long Fu
- grid.412524.40000 0004 0632 3994Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai, 200030 China
| | - Wen Yu
- grid.412524.40000 0004 0632 3994Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai, 200030 China
| |
Collapse
|
16
|
Lin G, Wang X, Ye H, Cao W. Radiomic Models Predict Tumor Microenvironment Using Artificial Intelligence-the Novel Biomarkers in Breast Cancer Immune Microenvironment. Technol Cancer Res Treat 2023; 22:15330338231218227. [PMID: 38111330 PMCID: PMC10734346 DOI: 10.1177/15330338231218227] [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: 09/12/2023] [Revised: 10/22/2023] [Accepted: 11/16/2023] [Indexed: 12/20/2023] Open
Abstract
Breast cancer is the most common malignancy in women, and some subtypes are associated with a poor prognosis with a lack of efficacious therapy. Moreover, immunotherapy and the use of other novel antibody‒drug conjugates have been rapidly incorporated into the standard management of advanced breast cancer. To extract more benefit from these therapies, clarifying and monitoring the tumor microenvironment (TME) status is critical, but this is difficult to accomplish based on conventional approaches. Radiomics is a method wherein radiological image features are comprehensively collected and assessed to build connections with disease diagnosis, prognosis, therapy efficacy, the TME, etc In recent years, studies focused on predicting the TME using radiomics have increasingly emerged, most of which demonstrate meaningful results and show better capability than conventional methods in some aspects. Beyond predicting tumor-infiltrating lymphocytes, immunophenotypes, cytokines, infiltrating inflammatory factors, and other stromal components, radiomic models have the potential to provide a completely new approach to deciphering the TME and facilitating tumor management by physicians.
Collapse
Affiliation(s)
- Guang Lin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xiaojia Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Hunan Ye
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Wenming Cao
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
17
|
Laurent PA, Morel D, Meziani L, Depil S, Deutsch E. Radiotherapy as a means to increase the efficacy of T-cell therapy in solid tumors. Oncoimmunology 2022; 12:2158013. [PMID: 36567802 PMCID: PMC9788698 DOI: 10.1080/2162402x.2022.2158013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Chimeric antigen receptor (CAR)-T cells have demonstrated significant improvements in the treatment of refractory B-cell malignancies that previously showed limited survival. In contrast, early-phase clinical studies targeting solid tumors have been disappointing. This may be due to both a lack of specific and homogeneously expressed targets at the surface of tumor cells, as well as intrinsic properties of the solid tumor microenvironment that limit homing and activation of adoptive T cells. Faced with these antagonistic conditions, radiotherapy (RT) has the potential to change the overall tumor landscape, from depleting tumor cells to reshaping the tumor microenvironment. In this article, we describe the current landscape and discuss how RT may play a pivotal role for enhancing the efficacy of adoptive T-cell therapies in solid tumors. Indeed, by improving homing, expansion and activation of infused T cells while reducing tumor volume and heterogeneity, the use of RT could help the implementation of engineered T cells in the treatment of solid tumors.
Collapse
Affiliation(s)
- Pierre-Antoine Laurent
- Department of Radiation Oncology, Gustave Roussy Cancer Campus; UNICANCER, Villejuif, France
- INSERM U1030, Molecular Radiation Therapy and Therapeutic Innovation, Gustave Roussy Cancer Campus, University of Paris-Saclay, SIRIC SOCRATE, Villejuif, France
| | - Daphne Morel
- Drug Development Department (D.I.T.E.P), Gustave Roussy Cancer Campus; UNICANCER, Villejuif, France
| | - Lydia Meziani
- INSERM U1030, Molecular Radiation Therapy and Therapeutic Innovation, Gustave Roussy Cancer Campus, University of Paris-Saclay, SIRIC SOCRATE, Villejuif, France
| | | | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy Cancer Campus; UNICANCER, Villejuif, France
- INSERM U1030, Molecular Radiation Therapy and Therapeutic Innovation, Gustave Roussy Cancer Campus, University of Paris-Saclay, SIRIC SOCRATE, Villejuif, France
| |
Collapse
|
18
|
Zhu Z, Chen M, Hu G, Pan Z, Han W, Tan W, Zhou Z, Wang M, Mao L, Li X, Sui X, Song L, Xu Y, Song W, Yu Y, Jin Z. A pre-treatment CT-based weighted radiomic approach combined with clinical characteristics to predict durable clinical benefits of immunotherapy in advanced lung cancer. Eur Radiol 2022; 33:3918-3930. [PMID: 36515714 PMCID: PMC9748402 DOI: 10.1007/s00330-022-09337-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/08/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To develop a pre-treatment CT-based predictive model to anticipate inoperable lung cancer patients' progression-free survival (PFS) to immunotherapy. METHODS This single-center retrospective study developed and cross-validated a radiomic model in 185 patients and tested it in 48 patients. The binary endpoint is the durable clinical benefit (DCB, PFS ≥ 6 months) and non-DCB (NDCB, PFS < 6 months). Radiomic features were extracted from multiple intrapulmonary lesions and weighted by an attention-based multiple-instance learning model. Aggregated features were then selected through L2-regularized ridge regression. Five machine-learning classifiers were conducted to build predictive models using radiomic and clinical features alone and then together. Lastly, the predictive value of the model with the best performance was validated by Kaplan-Meier survival analysis. RESULTS The predictive models based on the weighted radiomic approach showed superior performance across all classifiers (AUCs: 0.75-0.82) compared with the largest lesion approach (AUCs: 0.70-0.78) and the average sum approach (AUCs: 0.64-0.80). Among them, the logistic regression model yielded the most balanced performance (AUC = 0.87 [95%CI 0.84-0.89], 0.75 [0.68-0.82], 0.80 [0.68-0.92] in the training, validation, and test cohort respectively). The addition of five clinical characteristics significantly enhanced the performance of radiomic-only model (train: AUC 0.91 [0.89-0.93], p = .042; validation: AUC 0.86 [0.80-0.91], p = .011; test: AUC 0.86 [0.76-0.96], p = .026). Kaplan-Meier analysis of the radiomic-based predictive models showed a clear stratification between classifier-predicted DCB versus NDCB for PFS (HR = 2.40-2.95, p < 0.05). CONCLUSIONS The adoption of weighted radiomic features from multiple intrapulmonary lesions has the potential to predict long-term PFS benefits for patients who are candidates for PD-1/PD-L1 immunotherapies. KEY POINTS • Weighted radiomic-based model derived from multiple intrapulmonary lesions on pre-treatment CT images has the potential to predict durable clinical benefits of immunotherapy in lung cancer. • Early line immunotherapy is associated with longer progression-free survival in advanced lung cancer.
Collapse
Affiliation(s)
- Zhenchen Zhu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China ,4+4 Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China
| | - Minjiang Chen
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Ge Hu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Zhengsong Pan
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China ,4+4 Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China
| | - Wei Han
- Department of Epidemiology and Biostatistics, Institute of Basic Medicine Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, 100005 China
| | - Weixiong Tan
- Deepwise Artificial Intelligence (AI) Lab, Beijing Deepwise & League of PhD technology Co. Ltd, Beijing, 100080 China
| | - Zhen Zhou
- Deepwise Artificial Intelligence (AI) Lab, Beijing Deepwise & League of PhD technology Co. Ltd, Beijing, 100080 China
| | - Mengzhao Wang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Li Mao
- Deepwise Artificial Intelligence (AI) Lab, Beijing Deepwise & League of PhD technology Co. Ltd, Beijing, 100080 China
| | - Xiuli Li
- Deepwise Artificial Intelligence (AI) Lab, Beijing Deepwise & League of PhD technology Co. Ltd, Beijing, 100080 China
| | - Xin Sui
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Lan Song
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Yan Xu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Wei Song
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Yizhou Yu
- Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | - Zhengyu Jin
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| |
Collapse
|
19
|
Sun R, Lerousseau M, Briend-Diop J, Routier E, Roy S, Henry T, Ka K, Jiang R, Temar N, Carré A, Laville A, Hamaoui A, Laurent PA, Rouyar A, Robert C, Robert C, Deutsch E. Radiomics to evaluate interlesion heterogeneity and to predict lesion response and patient outcomes using a validated signature of CD8 cells in advanced melanoma patients treated with anti-PD1 immunotherapy. J Immunother Cancer 2022; 10:jitc-2022-004867. [PMID: 36307149 PMCID: PMC9621183 DOI: 10.1136/jitc-2022-004867] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2022] [Indexed: 11/07/2022] Open
Abstract
PURPOSE While there is still a significant need to identify potential biomarkers that can predict which patients are most likely to respond to immunotherapy treatments, radiomic approaches have shown promising results. The objectives of this study were to evaluate whether a previously validated radiomics signature of CD8 T-cells could predict progressions at a lesion level and whether the spatial heterogeneity of this radiomics score could be used at a patient level to assess the clinical response and survival of melanoma patients. METHODS Clinical data from patients with advanced melanoma treated in our center with immunotherapy were retrieved. Radiomic features were extracted and the CD8 radiomics signature was applied. A progressive lesion was defined by an increase in lesion size of 20% or more. Dispersion metrics of the radiomics signature were estimated to evaluate the impact of interlesion heterogeneity on patient's response. Fine-tuned cut-offs for predicting overall survival were evaluated after splitting data into training and test sets. RESULTS A total of 136 patients were included in this study, with 1120 segmented lesions at baseline, and 1052 lesions at first evaluation. A low CD8 radiomics score at baseline was associated with a significantly higher risk of lesion progression (AUC=0.55, p=0.0091), especially for lesions larger than >1 mL (AUC=0.59 overall, p=0.0035, with AUC=0.75, p=0.002 for subcutaneous lesions, AUC=0.68, p=0.01, for liver lesions and AUC=0.62, p=0.03 for nodes). The least infiltrated lesion according to the radiomics score of CD8 T-cells was positively associated with overall survival (training set HR=0.31, p=0.00062, test set HR=0.28, p=0.016), which remained significant in a multivariate analysis including clinical and biological variables. CONCLUSIONS These results confirm the predictive value at a lesion level of the biologically inspired CD8 radiomics score in melanoma patients treated with anti-PD1-based immunotherapy and may be interesting to assess the disease spatial heterogeneity to evaluate the patient prognosis with potential clinical implication such as tumor selection for focal ablative therapies.
Collapse
Affiliation(s)
- Roger Sun
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France,Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| | - Marvin Lerousseau
- Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| | - Jade Briend-Diop
- Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| | - Emilie Routier
- Dermatology Unit, Department of Medicine, Gustave Roussy, Villejuif, France,Université Paris Saclay, Inserm U981, Prédicteurs moléculaires et nouvelles cibles en oncologie, Gustave Roussy, Villejuif, France
| | - Severine Roy
- Dermatology Unit, Department of Medicine, Gustave Roussy, Villejuif, France,Université Paris Saclay, Inserm U981, Prédicteurs moléculaires et nouvelles cibles en oncologie, Gustave Roussy, Villejuif, France
| | - Théophraste Henry
- Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France,Imaging Department, Gustave Roussy, Villejuif, France
| | - Kanta Ka
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
| | - Rui Jiang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Nawal Temar
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
| | - Alexandre Carré
- Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| | - Adrien Laville
- Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| | - Anthony Hamaoui
- Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| | - Pierre-Antoine Laurent
- Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| | - Angela Rouyar
- Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| | - Charlotte Robert
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France,Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| | - Caroline Robert
- Dermatology Unit, Department of Medicine, Gustave Roussy, Villejuif, France,Université Paris Saclay, Inserm U981, Prédicteurs moléculaires et nouvelles cibles en oncologie, Gustave Roussy, Villejuif, France
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France,Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| |
Collapse
|