6
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Integration of longitudinal deep-radiomics and clinical data improves the prediction of durable benefits to anti-PD-1/PD-L1 immunotherapy in advanced NSCLC patients

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          Identifying predictive non-invasive biomarkers of immunotherapy response is crucial to avoid premature treatment interruptions or ineffective prolongation. Our aim was to develop a non-invasive biomarker for predicting immunotherapy clinical durable benefit, based on the integration of radiomics and clinical data monitored through early anti-PD-1/PD-L1 monoclonal antibodies treatment in patients with advanced non-small cell lung cancer (NSCLC).

          Methods

          In this study, 264 patients with pathologically confirmed stage IV NSCLC treated with immunotherapy were retrospectively collected from two institutions. The cohort was randomly divided into a training (n = 221) and an independent test set (n = 43), ensuring the balanced availability of baseline and follow-up data for each patient. Clinical data corresponding to the start of treatment was retrieved from electronic patient records, and blood test variables after the first and third cycles of immunotherapy were also collected. Additionally, traditional radiomics and deep-radiomics features were extracted from the primary tumors of the computed tomography (CT) scans before treatment and during patient follow-up. Random Forest was used to implementing baseline and longitudinal models using clinical and radiomics data separately, and then an ensemble model was built integrating both sources of information.

          Results

          The integration of longitudinal clinical and deep-radiomics data significantly improved clinical durable benefit prediction at 6 and 9 months after treatment in the independent test set, achieving an area under the receiver operating characteristic curve of 0.824 (95% CI: [0.658,0.953]) and 0.753 (95% CI: [0.549,0.931]). The Kaplan-Meier survival analysis showed that, for both endpoints, the signatures significantly stratified high- and low-risk patients (p-value< 0.05) and were significantly correlated with progression-free survival (PFS6 model: C-index 0.723, p-value = 0.004; PFS9 model: C-index 0.685, p-value = 0.030) and overall survival (PFS6 models: C-index 0.768, p-value = 0.002; PFS9 model: C-index 0.736, p-value = 0.023).

          Conclusions

          Integrating multidimensional and longitudinal data improved clinical durable benefit prediction to immunotherapy treatment of advanced non-small cell lung cancer patients. The selection of effective treatment and the appropriate evaluation of clinical benefit are important for better managing cancer patients with prolonged survival and preserving quality of life.

          Supplementary Information

          The online version contains supplementary material available at 10.1186/s12967-023-04004-x.

          Related collections

          Most cited references34

          • Record: found
          • Abstract: found
          • Article: not found

          New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1).

          Assessment of the change in tumour burden is an important feature of the clinical evaluation of cancer therapeutics: both tumour shrinkage (objective response) and disease progression are useful endpoints in clinical trials. Since RECIST was published in 2000, many investigators, cooperative groups, industry and government authorities have adopted these criteria in the assessment of treatment outcomes. However, a number of questions and issues have arisen which have led to the development of a revised RECIST guideline (version 1.1). Evidence for changes, summarised in separate papers in this special issue, has come from assessment of a large data warehouse (>6500 patients), simulation studies and literature reviews. HIGHLIGHTS OF REVISED RECIST 1.1: Major changes include: Number of lesions to be assessed: based on evidence from numerous trial databases merged into a data warehouse for analysis purposes, the number of lesions required to assess tumour burden for response determination has been reduced from a maximum of 10 to a maximum of five total (and from five to two per organ, maximum). Assessment of pathological lymph nodes is now incorporated: nodes with a short axis of 15 mm are considered measurable and assessable as target lesions. The short axis measurement should be included in the sum of lesions in calculation of tumour response. Nodes that shrink to <10mm short axis are considered normal. Confirmation of response is required for trials with response primary endpoint but is no longer required in randomised studies since the control arm serves as appropriate means of interpretation of data. Disease progression is clarified in several aspects: in addition to the previous definition of progression in target disease of 20% increase in sum, a 5mm absolute increase is now required as well to guard against over calling PD when the total sum is very small. Furthermore, there is guidance offered on what constitutes 'unequivocal progression' of non-measurable/non-target disease, a source of confusion in the original RECIST guideline. Finally, a section on detection of new lesions, including the interpretation of FDG-PET scan assessment is included. Imaging guidance: the revised RECIST includes a new imaging appendix with updated recommendations on the optimal anatomical assessment of lesions. A key question considered by the RECIST Working Group in developing RECIST 1.1 was whether it was appropriate to move from anatomic unidimensional assessment of tumour burden to either volumetric anatomical assessment or to functional assessment with PET or MRI. It was concluded that, at present, there is not sufficient standardisation or evidence to abandon anatomical assessment of tumour burden. The only exception to this is in the use of FDG-PET imaging as an adjunct to determination of progression. As is detailed in the final paper in this special issue, the use of these promising newer approaches requires appropriate clinical validation studies.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Radiomics: Images Are More than Pictures, They Are Data

            This report describes the process of radiomics, its challenges, and its potential power to facilitate better clinical decision making, particularly in the care of patients with cancer.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Computational Radiomics System to Decode the Radiographic Phenotype

              Radiomics aims to quantify phenotypic characteristics on medical imaging through the use of automated algorithms. Radiomic artificial intelligence (AI) technology, either based on engineered hard-coded algorithms or deep learning methods, can be used to develop non-invasive imaging-based biomarkers. However, lack of standardized algorithm definitions and image processing severely hampers reproducibility and comparability of results. To address this issue, we developed PyRadiomics , a flexible open-source platform capable of extracting a large panel of engineered features from medical images. PyRadiomics is implemented in Python and can be used standalone or using 3D-Slicer. Here, we discuss the workflow and architecture of PyRadiomics and demonstrate its application in characterizing lung-lesions. Source code, documentation, and examples are publicly available at www.radiomics.io . With this platform, we aim to establish a reference standard for radiomic analyses, provide a tested and maintained resource, and to grow the community of radiomic developers addressing critical needs in cancer research.
                Bookmark

                Author and article information

                Contributors
                benito.farina@upm.es
                mj.ledesma@upm.es
                Journal
                J Transl Med
                J Transl Med
                Journal of Translational Medicine
                BioMed Central (London )
                1479-5876
                5 March 2023
                5 March 2023
                2023
                : 21
                : 174
                Affiliations
                [1 ]GRID grid.5690.a, ISNI 0000 0001 2151 2978, Biomedical Image Technologies, ETSI Telecomunicación, Universidad Politécnica de Madrid, ; 28040 Madrid, Spain
                [2 ]GRID grid.429738.3, ISNI 0000 0004 1763 291X, Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), ; Madrid, Spain
                [3 ]GRID grid.419651.e, ISNI 0000 0000 9538 1950, Hospital Universitario Fundación Jiménez Díaz, ; 28040 Madrid, Spain
                [4 ]GRID grid.411730.0, ISNI 0000 0001 2191 685X, Clínica Universidad de Navarra, ; 28027 Madrid, Spain
                [5 ]GRID grid.512891.6, Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), ; Pamplona, Spain
                [6 ]GRID grid.510933.d, ISNI 0000 0004 8339 0058, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), ; 31008 Pamplona, Spain
                [7 ]GRID grid.7840.b, ISNI 0000 0001 2168 9183, Bioengineering Department, Universidad Carlos III de Madrid, ; 28911 Leganés, Spain
                [8 ]GRID grid.410526.4, ISNI 0000 0001 0277 7938, Instituto de Investigación Sanitaria Gregorio Marañón, ; 28007 Madrid, Spain
                [9 ]GRID grid.411730.0, ISNI 0000 0001 2191 685X, Department of Oncology, , Clínica Universidad de Navarra, ; 31008 Pamplona, Spain
                [10 ]GRID grid.5924.a, ISNI 0000000419370271, Program in Solid Tumors, Center for Applied Medical Research (CIMA), ; 31008 Pamplona, Spain
                [11 ]GRID grid.508840.1, ISNI 0000 0004 7662 6114, Navarra Institute for Health Research, IdiSNA, ; 31008 Pamplona, Spain
                [12 ]GRID grid.418082.7, ISNI 0000 0004 1771 144X, Department of Oncology, , Fundación Instituto Valenciano de Oncología (FIVO), ; 46009 Valencia, Spain
                Author information
                http://orcid.org/0000-0003-1674-5907
                Article
                4004
                10.1186/s12967-023-04004-x
                9985838
                36872371
                4619fb89-5e03-46bd-a3a1-09065717512a
                © The Author(s) 2023

                Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

                History
                : 30 November 2022
                : 16 February 2023
                Categories
                Research
                Custom metadata
                © The Author(s) 2023

                Medicine
                immunotherapy,lung cancer,clinical durable benefit,deep-radiomics,clinical data,longitudinal analysis,treatment monitoring

                Comments

                Comment on this article