1
|
Prior O, Macarro C, Navarro V, Monreal C, Ligero M, Garcia-Ruiz A, Serna G, Simonetti S, Braña I, Vieito M, Escobar M, Capdevila J, Byrne AT, Dienstmann R, Toledo R, Nuciforo P, Garralda E, Grussu F, Bernatowicz K, Perez-Lopez R. Identification of Precise 3D CT Radiomics for Habitat Computation by Machine Learning in Cancer. Radiol Artif Intell 2024; 6:e230118. [PMID: 38294307 PMCID: PMC10982821 DOI: 10.1148/ryai.230118] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 10/18/2023] [Accepted: 01/07/2024] [Indexed: 02/01/2024]
Abstract
Purpose To identify precise three-dimensional radiomics features in CT images that enable computation of stable and biologically meaningful habitats with machine learning for cancer heterogeneity assessment. Materials and Methods This retrospective study included 2436 liver or lung lesions from 605 CT scans (November 2010-December 2021) in 331 patients with cancer (mean age, 64.5 years ± 10.1 [SD]; 185 male patients). Three-dimensional radiomics were computed from original and perturbed (simulated retest) images with different combinations of feature computation kernel radius and bin size. The lower 95% confidence limit (LCL) of the intraclass correlation coefficient (ICC) was used to measure repeatability and reproducibility. Precise features were identified by combining repeatability and reproducibility results (LCL of ICC ≥ 0.50). Habitats were obtained with Gaussian mixture models in original and perturbed data using precise radiomics features and compared with habitats obtained using all features. The Dice similarity coefficient (DSC) was used to assess habitat stability. Biologic correlates of CT habitats were explored in a case study, with a cohort of 13 patients with CT, multiparametric MRI, and tumor biopsies. Results Three-dimensional radiomics showed poor repeatability (LCL of ICC: median [IQR], 0.442 [0.312-0.516]) and poor reproducibility against kernel radius (LCL of ICC: median [IQR], 0.440 [0.33-0.526]) but excellent reproducibility against bin size (LCL of ICC: median [IQR], 0.929 [0.853-0.988]). Twenty-six radiomics features were precise, differing in lung and liver lesions. Habitats obtained with precise features (DSC: median [IQR], 0.601 [0.494-0.712] and 0.651 [0.52-0.784] for lung and liver lesions, respectively) were more stable than those obtained with all features (DSC: median [IQR], 0.532 [0.424-0.637] and 0.587 [0.465-0.703] for lung and liver lesions, respectively; P < .001). In the case study, CT habitats correlated quantitatively and qualitatively with heterogeneity observed in multiparametric MRI habitats and histology. Conclusion Precise three-dimensional radiomics features were identified on CT images that enabled tumor heterogeneity assessment through stable tumor habitat computation. Keywords: CT, Diffusion-weighted Imaging, Dynamic Contrast-enhanced MRI, MRI, Radiomics, Unsupervised Learning, Oncology, Liver, Lung Supplemental material is available for this article. © RSNA, 2024 See also the commentary by Sagreiya in this issue.
Collapse
Affiliation(s)
- Olivia Prior
- From the Radiomics Group, Vall d’Hebron Institute of Oncology,
Carrer de Natzaret 115–117, Barcelona 08035, Spain (O.P., C. Macarro, C.
Monreal, M.L., A.G.R., F.G., K.B., R.P.L.); Oncology Data Science Group, Vall
d’Hebron Institute of Oncology, Barcelona, Spain (V.N., R.D.); Molecular
Pathology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
(G.S., S.S., P.N.); Department of Medical Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain (I.B., M.V., E.G., J.C.); Molecular
Therapeutic Research Unit, Vall d’Hebron Institute of Oncology,
Barcelona, Spain (I.B., M.V., E.G., J.C.); Department of Radiology, Vall
d’Hebron University Hospital, Barcelona, Spain (M.E.); Biomakers and
Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO),
Barcelona, Spain (R.T.); Department of Physiology and Medical Physics, Centre
for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
(A.T.B.); and National Pre-clinical Imaging Centre, Dublin, Ireland
(A.T.B.)
| | - Carlos Macarro
- From the Radiomics Group, Vall d’Hebron Institute of Oncology,
Carrer de Natzaret 115–117, Barcelona 08035, Spain (O.P., C. Macarro, C.
Monreal, M.L., A.G.R., F.G., K.B., R.P.L.); Oncology Data Science Group, Vall
d’Hebron Institute of Oncology, Barcelona, Spain (V.N., R.D.); Molecular
Pathology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
(G.S., S.S., P.N.); Department of Medical Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain (I.B., M.V., E.G., J.C.); Molecular
Therapeutic Research Unit, Vall d’Hebron Institute of Oncology,
Barcelona, Spain (I.B., M.V., E.G., J.C.); Department of Radiology, Vall
d’Hebron University Hospital, Barcelona, Spain (M.E.); Biomakers and
Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO),
Barcelona, Spain (R.T.); Department of Physiology and Medical Physics, Centre
for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
(A.T.B.); and National Pre-clinical Imaging Centre, Dublin, Ireland
(A.T.B.)
| | - Víctor Navarro
- From the Radiomics Group, Vall d’Hebron Institute of Oncology,
Carrer de Natzaret 115–117, Barcelona 08035, Spain (O.P., C. Macarro, C.
Monreal, M.L., A.G.R., F.G., K.B., R.P.L.); Oncology Data Science Group, Vall
d’Hebron Institute of Oncology, Barcelona, Spain (V.N., R.D.); Molecular
Pathology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
(G.S., S.S., P.N.); Department of Medical Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain (I.B., M.V., E.G., J.C.); Molecular
Therapeutic Research Unit, Vall d’Hebron Institute of Oncology,
Barcelona, Spain (I.B., M.V., E.G., J.C.); Department of Radiology, Vall
d’Hebron University Hospital, Barcelona, Spain (M.E.); Biomakers and
Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO),
Barcelona, Spain (R.T.); Department of Physiology and Medical Physics, Centre
for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
(A.T.B.); and National Pre-clinical Imaging Centre, Dublin, Ireland
(A.T.B.)
| | - Camilo Monreal
- From the Radiomics Group, Vall d’Hebron Institute of Oncology,
Carrer de Natzaret 115–117, Barcelona 08035, Spain (O.P., C. Macarro, C.
Monreal, M.L., A.G.R., F.G., K.B., R.P.L.); Oncology Data Science Group, Vall
d’Hebron Institute of Oncology, Barcelona, Spain (V.N., R.D.); Molecular
Pathology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
(G.S., S.S., P.N.); Department of Medical Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain (I.B., M.V., E.G., J.C.); Molecular
Therapeutic Research Unit, Vall d’Hebron Institute of Oncology,
Barcelona, Spain (I.B., M.V., E.G., J.C.); Department of Radiology, Vall
d’Hebron University Hospital, Barcelona, Spain (M.E.); Biomakers and
Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO),
Barcelona, Spain (R.T.); Department of Physiology and Medical Physics, Centre
for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
(A.T.B.); and National Pre-clinical Imaging Centre, Dublin, Ireland
(A.T.B.)
| | - Marta Ligero
- From the Radiomics Group, Vall d’Hebron Institute of Oncology,
Carrer de Natzaret 115–117, Barcelona 08035, Spain (O.P., C. Macarro, C.
Monreal, M.L., A.G.R., F.G., K.B., R.P.L.); Oncology Data Science Group, Vall
d’Hebron Institute of Oncology, Barcelona, Spain (V.N., R.D.); Molecular
Pathology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
(G.S., S.S., P.N.); Department of Medical Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain (I.B., M.V., E.G., J.C.); Molecular
Therapeutic Research Unit, Vall d’Hebron Institute of Oncology,
Barcelona, Spain (I.B., M.V., E.G., J.C.); Department of Radiology, Vall
d’Hebron University Hospital, Barcelona, Spain (M.E.); Biomakers and
Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO),
Barcelona, Spain (R.T.); Department of Physiology and Medical Physics, Centre
for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
(A.T.B.); and National Pre-clinical Imaging Centre, Dublin, Ireland
(A.T.B.)
| | - Alonso Garcia-Ruiz
- From the Radiomics Group, Vall d’Hebron Institute of Oncology,
Carrer de Natzaret 115–117, Barcelona 08035, Spain (O.P., C. Macarro, C.
Monreal, M.L., A.G.R., F.G., K.B., R.P.L.); Oncology Data Science Group, Vall
d’Hebron Institute of Oncology, Barcelona, Spain (V.N., R.D.); Molecular
Pathology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
(G.S., S.S., P.N.); Department of Medical Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain (I.B., M.V., E.G., J.C.); Molecular
Therapeutic Research Unit, Vall d’Hebron Institute of Oncology,
Barcelona, Spain (I.B., M.V., E.G., J.C.); Department of Radiology, Vall
d’Hebron University Hospital, Barcelona, Spain (M.E.); Biomakers and
Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO),
Barcelona, Spain (R.T.); Department of Physiology and Medical Physics, Centre
for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
(A.T.B.); and National Pre-clinical Imaging Centre, Dublin, Ireland
(A.T.B.)
| | - Garazi Serna
- From the Radiomics Group, Vall d’Hebron Institute of Oncology,
Carrer de Natzaret 115–117, Barcelona 08035, Spain (O.P., C. Macarro, C.
Monreal, M.L., A.G.R., F.G., K.B., R.P.L.); Oncology Data Science Group, Vall
d’Hebron Institute of Oncology, Barcelona, Spain (V.N., R.D.); Molecular
Pathology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
(G.S., S.S., P.N.); Department of Medical Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain (I.B., M.V., E.G., J.C.); Molecular
Therapeutic Research Unit, Vall d’Hebron Institute of Oncology,
Barcelona, Spain (I.B., M.V., E.G., J.C.); Department of Radiology, Vall
d’Hebron University Hospital, Barcelona, Spain (M.E.); Biomakers and
Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO),
Barcelona, Spain (R.T.); Department of Physiology and Medical Physics, Centre
for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
(A.T.B.); and National Pre-clinical Imaging Centre, Dublin, Ireland
(A.T.B.)
| | - Sara Simonetti
- From the Radiomics Group, Vall d’Hebron Institute of Oncology,
Carrer de Natzaret 115–117, Barcelona 08035, Spain (O.P., C. Macarro, C.
Monreal, M.L., A.G.R., F.G., K.B., R.P.L.); Oncology Data Science Group, Vall
d’Hebron Institute of Oncology, Barcelona, Spain (V.N., R.D.); Molecular
Pathology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
(G.S., S.S., P.N.); Department of Medical Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain (I.B., M.V., E.G., J.C.); Molecular
Therapeutic Research Unit, Vall d’Hebron Institute of Oncology,
Barcelona, Spain (I.B., M.V., E.G., J.C.); Department of Radiology, Vall
d’Hebron University Hospital, Barcelona, Spain (M.E.); Biomakers and
Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO),
Barcelona, Spain (R.T.); Department of Physiology and Medical Physics, Centre
for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
(A.T.B.); and National Pre-clinical Imaging Centre, Dublin, Ireland
(A.T.B.)
| | - Irene Braña
- From the Radiomics Group, Vall d’Hebron Institute of Oncology,
Carrer de Natzaret 115–117, Barcelona 08035, Spain (O.P., C. Macarro, C.
Monreal, M.L., A.G.R., F.G., K.B., R.P.L.); Oncology Data Science Group, Vall
d’Hebron Institute of Oncology, Barcelona, Spain (V.N., R.D.); Molecular
Pathology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
(G.S., S.S., P.N.); Department of Medical Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain (I.B., M.V., E.G., J.C.); Molecular
Therapeutic Research Unit, Vall d’Hebron Institute of Oncology,
Barcelona, Spain (I.B., M.V., E.G., J.C.); Department of Radiology, Vall
d’Hebron University Hospital, Barcelona, Spain (M.E.); Biomakers and
Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO),
Barcelona, Spain (R.T.); Department of Physiology and Medical Physics, Centre
for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
(A.T.B.); and National Pre-clinical Imaging Centre, Dublin, Ireland
(A.T.B.)
| | - Maria Vieito
- From the Radiomics Group, Vall d’Hebron Institute of Oncology,
Carrer de Natzaret 115–117, Barcelona 08035, Spain (O.P., C. Macarro, C.
Monreal, M.L., A.G.R., F.G., K.B., R.P.L.); Oncology Data Science Group, Vall
d’Hebron Institute of Oncology, Barcelona, Spain (V.N., R.D.); Molecular
Pathology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
(G.S., S.S., P.N.); Department of Medical Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain (I.B., M.V., E.G., J.C.); Molecular
Therapeutic Research Unit, Vall d’Hebron Institute of Oncology,
Barcelona, Spain (I.B., M.V., E.G., J.C.); Department of Radiology, Vall
d’Hebron University Hospital, Barcelona, Spain (M.E.); Biomakers and
Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO),
Barcelona, Spain (R.T.); Department of Physiology and Medical Physics, Centre
for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
(A.T.B.); and National Pre-clinical Imaging Centre, Dublin, Ireland
(A.T.B.)
| | - Manuel Escobar
- From the Radiomics Group, Vall d’Hebron Institute of Oncology,
Carrer de Natzaret 115–117, Barcelona 08035, Spain (O.P., C. Macarro, C.
Monreal, M.L., A.G.R., F.G., K.B., R.P.L.); Oncology Data Science Group, Vall
d’Hebron Institute of Oncology, Barcelona, Spain (V.N., R.D.); Molecular
Pathology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
(G.S., S.S., P.N.); Department of Medical Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain (I.B., M.V., E.G., J.C.); Molecular
Therapeutic Research Unit, Vall d’Hebron Institute of Oncology,
Barcelona, Spain (I.B., M.V., E.G., J.C.); Department of Radiology, Vall
d’Hebron University Hospital, Barcelona, Spain (M.E.); Biomakers and
Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO),
Barcelona, Spain (R.T.); Department of Physiology and Medical Physics, Centre
for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
(A.T.B.); and National Pre-clinical Imaging Centre, Dublin, Ireland
(A.T.B.)
| | - Jaume Capdevila
- From the Radiomics Group, Vall d’Hebron Institute of Oncology,
Carrer de Natzaret 115–117, Barcelona 08035, Spain (O.P., C. Macarro, C.
Monreal, M.L., A.G.R., F.G., K.B., R.P.L.); Oncology Data Science Group, Vall
d’Hebron Institute of Oncology, Barcelona, Spain (V.N., R.D.); Molecular
Pathology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
(G.S., S.S., P.N.); Department of Medical Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain (I.B., M.V., E.G., J.C.); Molecular
Therapeutic Research Unit, Vall d’Hebron Institute of Oncology,
Barcelona, Spain (I.B., M.V., E.G., J.C.); Department of Radiology, Vall
d’Hebron University Hospital, Barcelona, Spain (M.E.); Biomakers and
Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO),
Barcelona, Spain (R.T.); Department of Physiology and Medical Physics, Centre
for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
(A.T.B.); and National Pre-clinical Imaging Centre, Dublin, Ireland
(A.T.B.)
| | - Annette T. Byrne
- From the Radiomics Group, Vall d’Hebron Institute of Oncology,
Carrer de Natzaret 115–117, Barcelona 08035, Spain (O.P., C. Macarro, C.
Monreal, M.L., A.G.R., F.G., K.B., R.P.L.); Oncology Data Science Group, Vall
d’Hebron Institute of Oncology, Barcelona, Spain (V.N., R.D.); Molecular
Pathology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
(G.S., S.S., P.N.); Department of Medical Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain (I.B., M.V., E.G., J.C.); Molecular
Therapeutic Research Unit, Vall d’Hebron Institute of Oncology,
Barcelona, Spain (I.B., M.V., E.G., J.C.); Department of Radiology, Vall
d’Hebron University Hospital, Barcelona, Spain (M.E.); Biomakers and
Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO),
Barcelona, Spain (R.T.); Department of Physiology and Medical Physics, Centre
for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
(A.T.B.); and National Pre-clinical Imaging Centre, Dublin, Ireland
(A.T.B.)
| | - Rodrigo Dienstmann
- From the Radiomics Group, Vall d’Hebron Institute of Oncology,
Carrer de Natzaret 115–117, Barcelona 08035, Spain (O.P., C. Macarro, C.
Monreal, M.L., A.G.R., F.G., K.B., R.P.L.); Oncology Data Science Group, Vall
d’Hebron Institute of Oncology, Barcelona, Spain (V.N., R.D.); Molecular
Pathology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
(G.S., S.S., P.N.); Department of Medical Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain (I.B., M.V., E.G., J.C.); Molecular
Therapeutic Research Unit, Vall d’Hebron Institute of Oncology,
Barcelona, Spain (I.B., M.V., E.G., J.C.); Department of Radiology, Vall
d’Hebron University Hospital, Barcelona, Spain (M.E.); Biomakers and
Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO),
Barcelona, Spain (R.T.); Department of Physiology and Medical Physics, Centre
for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
(A.T.B.); and National Pre-clinical Imaging Centre, Dublin, Ireland
(A.T.B.)
| | - Rodrigo Toledo
- From the Radiomics Group, Vall d’Hebron Institute of Oncology,
Carrer de Natzaret 115–117, Barcelona 08035, Spain (O.P., C. Macarro, C.
Monreal, M.L., A.G.R., F.G., K.B., R.P.L.); Oncology Data Science Group, Vall
d’Hebron Institute of Oncology, Barcelona, Spain (V.N., R.D.); Molecular
Pathology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
(G.S., S.S., P.N.); Department of Medical Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain (I.B., M.V., E.G., J.C.); Molecular
Therapeutic Research Unit, Vall d’Hebron Institute of Oncology,
Barcelona, Spain (I.B., M.V., E.G., J.C.); Department of Radiology, Vall
d’Hebron University Hospital, Barcelona, Spain (M.E.); Biomakers and
Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO),
Barcelona, Spain (R.T.); Department of Physiology and Medical Physics, Centre
for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
(A.T.B.); and National Pre-clinical Imaging Centre, Dublin, Ireland
(A.T.B.)
| | - Paolo Nuciforo
- From the Radiomics Group, Vall d’Hebron Institute of Oncology,
Carrer de Natzaret 115–117, Barcelona 08035, Spain (O.P., C. Macarro, C.
Monreal, M.L., A.G.R., F.G., K.B., R.P.L.); Oncology Data Science Group, Vall
d’Hebron Institute of Oncology, Barcelona, Spain (V.N., R.D.); Molecular
Pathology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
(G.S., S.S., P.N.); Department of Medical Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain (I.B., M.V., E.G., J.C.); Molecular
Therapeutic Research Unit, Vall d’Hebron Institute of Oncology,
Barcelona, Spain (I.B., M.V., E.G., J.C.); Department of Radiology, Vall
d’Hebron University Hospital, Barcelona, Spain (M.E.); Biomakers and
Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO),
Barcelona, Spain (R.T.); Department of Physiology and Medical Physics, Centre
for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
(A.T.B.); and National Pre-clinical Imaging Centre, Dublin, Ireland
(A.T.B.)
| | - Elena Garralda
- From the Radiomics Group, Vall d’Hebron Institute of Oncology,
Carrer de Natzaret 115–117, Barcelona 08035, Spain (O.P., C. Macarro, C.
Monreal, M.L., A.G.R., F.G., K.B., R.P.L.); Oncology Data Science Group, Vall
d’Hebron Institute of Oncology, Barcelona, Spain (V.N., R.D.); Molecular
Pathology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
(G.S., S.S., P.N.); Department of Medical Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain (I.B., M.V., E.G., J.C.); Molecular
Therapeutic Research Unit, Vall d’Hebron Institute of Oncology,
Barcelona, Spain (I.B., M.V., E.G., J.C.); Department of Radiology, Vall
d’Hebron University Hospital, Barcelona, Spain (M.E.); Biomakers and
Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO),
Barcelona, Spain (R.T.); Department of Physiology and Medical Physics, Centre
for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
(A.T.B.); and National Pre-clinical Imaging Centre, Dublin, Ireland
(A.T.B.)
| | - Francesco Grussu
- From the Radiomics Group, Vall d’Hebron Institute of Oncology,
Carrer de Natzaret 115–117, Barcelona 08035, Spain (O.P., C. Macarro, C.
Monreal, M.L., A.G.R., F.G., K.B., R.P.L.); Oncology Data Science Group, Vall
d’Hebron Institute of Oncology, Barcelona, Spain (V.N., R.D.); Molecular
Pathology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
(G.S., S.S., P.N.); Department of Medical Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain (I.B., M.V., E.G., J.C.); Molecular
Therapeutic Research Unit, Vall d’Hebron Institute of Oncology,
Barcelona, Spain (I.B., M.V., E.G., J.C.); Department of Radiology, Vall
d’Hebron University Hospital, Barcelona, Spain (M.E.); Biomakers and
Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO),
Barcelona, Spain (R.T.); Department of Physiology and Medical Physics, Centre
for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
(A.T.B.); and National Pre-clinical Imaging Centre, Dublin, Ireland
(A.T.B.)
| | | | | |
Collapse
|
2
|
Chaika M, Männlin S, Gassenmaier S, Tsiflikas I, Dittmann H, Flaadt T, Warmann S, Gückel B, Schäfer JF. Combined Metabolic and Functional Tumor Volumes on [ 18F]FDG-PET/MRI in Neuroblastoma Using Voxel-Wise Analysis. J Clin Med 2023; 12:5976. [PMID: 37762918 PMCID: PMC10531552 DOI: 10.3390/jcm12185976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
PURPOSE The purpose of our study was to evaluate the association between the [18F]FDG standard uptake value (SUV) and the apparent diffusion coefficient (ADC) in neuroblastoma (NB) by voxel-wise analysis. METHODS From our prospective observational PET/MRI study, a subcohort of patients diagnosed with NB with both baseline imaging and post-chemotherapy imaging was further investigated. After registration and tumor segmentation, metabolic and functional tumor volumes were calculated from the ADC and SUV values using dedicated software allowing for voxel-wise analysis. Under the mean of thresholds, each voxel was assigned to one of three virtual tissue groups: highly vital (v) (low ADC and high SUV), possibly low vital (lv) (high ADC and low SUV), and equivocal (e) with high ADC and high SUV or low ADC and low SUV. Moreover, three clusters were generated from the total tumor volumes using the method of multiple Gaussian distributions. The Pearson's correlation coefficient between the ADC and the SUV was calculated for each group. RESULTS Out of 43 PET/MRIs in 21 patients with NB, 16 MRIs in 8 patients met the inclusion criteria (PET/MRIs before and after chemotherapy). The proportion of tumor volumes were 26%, 36%, and 38% (v, lv, e) at baseline, 0.03%, 66%, and 34% after treatment in patients with response, and 42%, 25%, and 33% with progressive disease, respectively. In all clusters, the ADC and the SUV correlated negatively. In the cluster that corresponded to highly vital tissue, the ADC and the SUV showed a moderate negative correlation before treatment (R = -0.18; p < 0.0001) and the strongest negative correlation after treatment (R = -0.45; p < 0.0001). Interestingly, only patients with progression (n = 2) under therapy had a relevant part in this cluster post-treatment. CONCLUSION Our results indicate that voxel-wise analysis of the ADC and the SUV is feasible and can quantify the different quality of tissue in neuroblastic tumors. Monitoring ADCs as well as SUV levels can quantify tumor dynamics during therapy.
Collapse
Affiliation(s)
- Maryanna Chaika
- Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Simon Männlin
- Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Sebastian Gassenmaier
- Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Ilias Tsiflikas
- Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Helmut Dittmann
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Tim Flaadt
- Department of Hematology and Oncology, University Children’s Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Steven Warmann
- Department of Pediatric Surgery and Pediatric Urology, University Children’s Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Brigitte Gückel
- Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, 72076 Tuebingen, Germany
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Jürgen Frank Schäfer
- Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, 72076 Tuebingen, Germany
| |
Collapse
|
3
|
Katiyar P, Schwenck J, Frauenfeld L, Divine MR, Agrawal V, Kohlhofer U, Gatidis S, Kontermann R, Königsrainer A, Quintanilla-Martinez L, la Fougère C, Schölkopf B, Pichler BJ, Disselhorst JA. Quantification of intratumoural heterogeneity in mice and patients via machine-learning models trained on PET-MRI data. Nat Biomed Eng 2023; 7:1014-1027. [PMID: 37277483 DOI: 10.1038/s41551-023-01047-9] [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: 01/05/2021] [Accepted: 04/26/2023] [Indexed: 06/07/2023]
Abstract
In oncology, intratumoural heterogeneity is closely linked with the efficacy of therapy, and can be partially characterized via tumour biopsies. Here we show that intratumoural heterogeneity can be characterized spatially via phenotype-specific, multi-view learning classifiers trained with data from dynamic positron emission tomography (PET) and multiparametric magnetic resonance imaging (MRI). Classifiers trained with PET-MRI data from mice with subcutaneous colon cancer quantified phenotypic changes resulting from an apoptosis-inducing targeted therapeutic and provided biologically relevant probability maps of tumour-tissue subtypes. When applied to retrospective PET-MRI data of patients with liver metastases from colorectal cancer, the trained classifiers characterized intratumoural tissue subregions in agreement with tumour histology. The spatial characterization of intratumoural heterogeneity in mice and patients via multimodal, multiparametric imaging aided by machine-learning may facilitate applications in precision oncology.
Collapse
Affiliation(s)
- Prateek Katiyar
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Max Planck Institute for Intelligent Systems, Tübingen, Germany
| | - Johannes Schwenck
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image Guided and Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Nuclear Medicine and Clinical Molecular Imaging, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Leonie Frauenfeld
- Institute of Pathology and Neuropathology, Eberhard Karls University Tübingen and Comprehensive Cancer Center, University Hospital Tübingen, Tübingen, Germany
| | - Mathew R Divine
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Vaibhav Agrawal
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Max Planck Institute for Intelligent Systems, Tübingen, Germany
| | - Ursula Kohlhofer
- Institute of Pathology and Neuropathology, Eberhard Karls University Tübingen and Comprehensive Cancer Center, University Hospital Tübingen, Tübingen, Germany
| | - Sergios Gatidis
- Cluster of Excellence iFIT (EXC 2180) 'Image Guided and Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Radiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Roland Kontermann
- Institute of Cell Biology and Immunology, SRCSB, University of Stuttgart, Stuttgart, Germany
| | - Alfred Königsrainer
- Cluster of Excellence iFIT (EXC 2180) 'Image Guided and Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, Tübingen, Germany
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Leticia Quintanilla-Martinez
- Cluster of Excellence iFIT (EXC 2180) 'Image Guided and Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, Tübingen, Germany
- Institute of Pathology and Neuropathology, Eberhard Karls University Tübingen and Comprehensive Cancer Center, University Hospital Tübingen, Tübingen, Germany
| | - Christian la Fougère
- Cluster of Excellence iFIT (EXC 2180) 'Image Guided and Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Nuclear Medicine and Clinical Molecular Imaging, Eberhard Karls University Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bernhard Schölkopf
- Max Planck Institute for Intelligent Systems, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image Guided and Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, Tübingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) 'Image Guided and Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, Tübingen, Germany.
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Jonathan A Disselhorst
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image Guided and Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
4
|
Schwenck J, Sonanini D, Cotton JM, Rammensee HG, la Fougère C, Zender L, Pichler BJ. Advances in PET imaging of cancer. Nat Rev Cancer 2023:10.1038/s41568-023-00576-4. [PMID: 37258875 DOI: 10.1038/s41568-023-00576-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/17/2023] [Indexed: 06/02/2023]
Abstract
Molecular imaging has experienced enormous advancements in the areas of imaging technology, imaging probe and contrast development, and data quality, as well as machine learning-based data analysis. Positron emission tomography (PET) and its combination with computed tomography (CT) or magnetic resonance imaging (MRI) as a multimodality PET-CT or PET-MRI system offer a wealth of molecular, functional and morphological data with a single patient scan. Despite the recent technical advances and the availability of dozens of disease-specific contrast and imaging probes, only a few parameters, such as tumour size or the mean tracer uptake, are used for the evaluation of images in clinical practice. Multiparametric in vivo imaging data not only are highly quantitative but also can provide invaluable information about pathophysiology, receptor expression, metabolism, or morphological and functional features of tumours, such as pH, oxygenation or tissue density, as well as pharmacodynamic properties of drugs, to measure drug response with a contrast agent. It can further quantitatively map and spatially resolve the intertumoural and intratumoural heterogeneity, providing insights into tumour vulnerabilities for target-specific therapeutic interventions. Failure to exploit and integrate the full potential of such powerful imaging data may lead to a lost opportunity in which patients do not receive the best possible care. With the desire to implement personalized medicine in the cancer clinic, the full comprehensive diagnostic power of multiplexed imaging should be utilized.
Collapse
Affiliation(s)
- Johannes Schwenck
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
- Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
| | - Dominik Sonanini
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
- Medical Oncology and Pulmonology, Department of Internal Medicine, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Jonathan M Cotton
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
| | - Hans-Georg Rammensee
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
- Department of Immunology, IFIZ Institute for Cell Biology, Eberhard Karls University of Tübingen, Tübingen, Germany
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany
| | - Christian la Fougère
- Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany
| | - Lars Zender
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
- Medical Oncology and Pulmonology, Department of Internal Medicine, Eberhard Karls University of Tübingen, Tübingen, Germany
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany.
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany.
| |
Collapse
|
5
|
Lapin A, Perfahl H, Jain HV, Reuss M. Integrating a dynamic central metabolism model of cancer cells with a hybrid 3D multiscale model for vascular hepatocellular carcinoma growth. Sci Rep 2022; 12:12373. [PMID: 35858953 PMCID: PMC9300625 DOI: 10.1038/s41598-022-15767-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/29/2022] [Indexed: 11/09/2022] Open
Abstract
We develop here a novel modelling approach with the aim of closing the conceptual gap between tumour-level metabolic processes and the metabolic processes occurring in individual cancer cells. In particular, the metabolism in hepatocellular carcinoma derived cell lines (HEPG2 cells) has been well characterized but implementations of multiscale models integrating this known metabolism have not been previously reported. We therefore extend a previously published multiscale model of vascular tumour growth, and integrate it with an experimentally verified network of central metabolism in HEPG2 cells. This resultant combined model links spatially heterogeneous vascular tumour growth with known metabolic networks within tumour cells and accounts for blood flow, angiogenesis, vascular remodelling and nutrient/growth factor transport within a growing tumour, as well as the movement of, and interactions between normal and cancer cells. Model simulations report for the first time, predictions of spatially resolved time courses of core metabolites in HEPG2 cells. These simulations can be performed at a sufficient scale to incorporate clinically relevant features of different tumour systems using reasonable computational resources. Our results predict larger than expected temporal and spatial heterogeneity in the intracellular concentrations of glucose, oxygen, lactate pyruvate, f16bp and Acetyl-CoA. The integrated multiscale model developed here provides an ideal quantitative framework in which to study the relationship between dosage, timing, and scheduling of anti-neoplastic agents and the physiological effects of tumour metabolism at the cellular level. Such models, therefore, have the potential to inform treatment decisions when drug response is dependent on the metabolic state of individual cancer cells.
Collapse
Affiliation(s)
- Alexey Lapin
- Stuttgart Research Center Systems Biology, University Stuttgart, Stuttgart, Germany
- Institute of Chemical Process Engineering, University Stuttgart, Stuttgart, Germany
| | - Holger Perfahl
- Stuttgart Research Center Systems Biology, University Stuttgart, Stuttgart, Germany
| | - Harsh Vardhan Jain
- Department of Mathematics and Statistics, University of Minnesota Duluth, Duluth, MN, USA
| | - Matthias Reuss
- Stuttgart Research Center Systems Biology, University Stuttgart, Stuttgart, Germany.
| |
Collapse
|
6
|
Ahangari S, Littrup Andersen F, Liv Hansen N, Jakobi Nøttrup T, Berthelsen AK, Folsted Kallehauge J, Richter Vogelius I, Kjaer A, Espe Hansen A, Fischer BM. Multi-parametric PET/MRI for enhanced tumor characterization of patients with cervical cancer. Eur J Hybrid Imaging 2022; 6:7. [PMID: 35378619 PMCID: PMC8980118 DOI: 10.1186/s41824-022-00129-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/07/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Aim
The concept of personalized medicine has brought increased awareness to the importance of inter- and intra-tumor heterogeneity for cancer treatment. The aim of this study was to explore simultaneous multi-parametric PET/MRI prior to chemoradiotherapy for cervical cancer for characterization of tumors and tumor heterogeneity.
Methods
Ten patients with histologically proven primary cervical cancer were examined with multi-parametric 68Ga-NODAGA-E[c(RGDyK)]2-PET/MRI for radiation treatment planning after diagnostic 18F-FDG-PET/CT. Standardized uptake values (SUV) of RGD and FDG, diffusion weighted MRI and the derived apparent diffusion coefficient (ADC), and pharmacokinetic maps obtained from dynamic contrast-enhanced MRI with the Tofts model (iAUC60, Ktrans, ve, and kep) were included in the analysis. The spatial relation between functional imaging parameters in tumors was examined by a correlation analysis and joint histograms at the voxel level. The ability of multi-parametric imaging to identify tumor tissue classes was explored using an unsupervised 3D Gaussian mixture model-based cluster analysis.
Results
Functional MRI and PET of cervical cancers appeared heterogeneous both between patients and spatially within the tumors, and the relations between parameters varied strongly within the patient cohort. The strongest spatial correlation was observed between FDG uptake and ADC (median r = − 0.7). There was moderate voxel-wise correlation between RGD and FDG uptake, and weak correlations between all other modalities. Distinct relations between the ADC and RGD uptake as well as the ADC and FDG uptake were apparent in joint histograms. A cluster analysis using the combination of ADC, FDG and RGD uptake suggested tissue classes which could potentially relate to tumor sub-volumes.
Conclusion
A multi-parametric PET/MRI examination of patients with cervical cancer integrated with treatment planning and including estimation of angiogenesis and glucose metabolism as well as MRI diffusion and perfusion parameters is feasible. A combined analysis of functional imaging parameters indicates a potential of multi-parametric PET/MRI to contribute to a better characterization of tumor heterogeneity than the modalities alone. However, the study is based on small patient numbers and further studies are needed prior to the future design of individually adapted treatment approaches based on multi-parametric functional imaging.
Collapse
|
7
|
Use of an optimised enzyme/prodrug combination for Clostridia directed enzyme prodrug therapy induces a significant growth delay in necrotic tumours. Cancer Gene Ther 2022; 29:178-188. [PMID: 33558701 DOI: 10.1038/s41417-021-00296-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/20/2020] [Accepted: 01/12/2021] [Indexed: 01/30/2023]
Abstract
Necrosis is a typical histological feature of solid tumours that provides a selective environment for growth of the non-pathogenic anaerobic bacterium Clostridium sporogenes. Modest anti-tumour activity as a single agent encouraged the use of C. sporogenes as a vector to express therapeutic genes selectively in tumour tissue, a concept termed Clostridium Directed Enzyme Prodrug Therapy (CDEPT). Here, we examine the ability of a recently identified Neisseria meningitidis type I nitroreductase (NmeNTR) to metabolise the prodrug PR-104A in an in vivo model of CDEPT. Human HCT116 colon cancer cells stably over-expressing NmeNTR demonstrated significant sensitivity to PR-104A, the imaging agent EF5, and several nitro(hetero)cyclic anti-infective compounds. Chemical induction of necrosis in human H1299 xenografts by the vascular disrupting agent vadimezan promoted colonisation by NmeNTR-expressing C. sporogenes, and efficacy studies demonstrated moderate but significant anti-tumour activity of spores when compared to untreated controls. Inclusion of the pre-prodrug PR-104 into the treatment schedule provided significant additional activity, indicating proof-of-principle. Successful preclinical evaluation of a transferable gene that enables metabolism of both PET imaging agents (for vector visualisation) and prodrugs (for conditional enhancement of efficacy) is an important step towards the prospect of CDEPT entering clinical evaluation.
Collapse
|
8
|
Kim C, Cho HH, Choi JY, Franks TJ, Han J, Choi Y, Lee SH, Park H, Lee KS. Pleomorphic carcinoma of the lung: Prognostic models of semantic, radiomics and combined features from CT and PET/CT in 85 patients. Eur J Radiol Open 2021; 8:100351. [PMID: 34041307 PMCID: PMC8141891 DOI: 10.1016/j.ejro.2021.100351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/03/2021] [Accepted: 05/08/2021] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION To demonstrate semantic, radiomics, and the combined risk models related to the prognoses of pulmonary pleomorphic carcinomas (PCs). METHODS We included 85 patients (M:F = 71:14; age, 35-88 [mean, 63 years]) whose imaging features were divided into training (n = 60) and test (n = 25) sets. Nineteen semantic and 142 radiomics features related to tumors were computed. Semantic risk score (SRS) model was built using the Cox-least absolute shrinkage and selection operator (LASSO) approach. Radiomics risk score (RRS) from CT and PET features and combined risk score (CRS) adopting both semantic and radiomics features were also constructed. Risk groups were stratified by the median of the risk scores of the training set. Survival analysis was conducted with the Kaplan-Meier plots. RESULTS Of 85 PCs, adenocarcinoma was the most common epithelial component found in 63 (73 %) tumors. In SRS model, four features were stratified into high- and low-risk groups (HR, 4.119; concordance index ([C-index], 0.664) in the test set. In RRS model, five features helped improve the stratification (HR, 3.716; C-index, 0.591) and in CRS model, three features helped perform the best stratification (HR, 4.795; C-index, 0.617). The two significant features of CRS models were the SUVmax and the histogram feature of energy ([CT Firstorder Energy]). CONCLUSION In PCs of the lungs, the combined model leveraging semantic and radiomics features provides a better prognosis compared to using semantic and radiomics features separately. The high SUVmax of solid portion (CT Firstorder Energy) of tumors is associated with poor prognosis in lung PCs.
Collapse
Key Words
- C-index, Concordance index
- CRS, Combined risk score
- DL, Deep learning
- GCLM, Gray-level co-occurrence matrix
- HR, Hazard ration
- ICC, Intra-class correlation
- ISZM, Intensity size zone matrix
- KRAS, Kirsten rat sarcoma viral oncogene homolog
- LASSO, Least absolute shrinkage and selection operator
- LDA, Low density area
- Lung
- MRI, Magnetic resonance imaging
- MTV, Metabolic tumor volume
- Non-small cell carcinoma
- PC, Pleomorphic carcinoma
- PET/CT, Positron emission tomography/Computed tomography
- Pleomorphic carcinoma
- Prognosis
- ROI, Region of interest
- RRS, Radiomics risk score
- Radiomics
- SRS, Semantic risk score
- SUVavg, Average standardized uptake value
- SUVmax, Maximum standardized uptake value
- TLG, Total lesion glycolysis
- VOI, Volume of interest
Collapse
Affiliation(s)
- Chohee Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, South Korea
| | - Hwan-ho Cho
- Department of Electronic and Computer Engineering, Sungkyunkwan University, Suwon, South Korea
| | - Joon Young Choi
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, South Korea
| | - Teri J. Franks
- Department of Pulmonary and Mediastinal Pathology, Department of Defense, The Joint Pathology Center, Silver Spring, MD, USA
| | - Joungho Han
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, South Korea
| | - Yeonu Choi
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, South Korea
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, South Korea
| | - Hyunjin Park
- School of Electronic and Electrical Engineering, Sungkyunkwan University, Suwon, South Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, South Korea
| | - Kyung Soo Lee
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, South Korea
| |
Collapse
|
9
|
Ferreira Junior JR, Koenigkam-Santos M, Machado CVB, Faleiros MC, Correia NSC, Cipriano FEG, Fabro AT, de Azevedo-Marques PM. Radiomic analysis of lung cancer for the assessment of patient prognosis and intratumor heterogeneity. Radiol Bras 2021; 54:87-93. [PMID: 33854262 PMCID: PMC8029936 DOI: 10.1590/0100-3984.2019.0135] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objective To determine whether the radiomic features of lung lesions on computed tomography correlate with overall survival in lung cancer patients. Materials and Methods This was a retrospective study involving 101 consecutive patients with malignant neoplasms confirmed by biopsy or surgery. On computed tomography images, the lesions were submitted to semi-automated segmentation and were characterized on the basis of 2,465 radiomic variables. The prognostic assessment was based on Kaplan-Meier analysis and log-rank tests, according to the median value of the radiomic variables. Results Of the 101 patients evaluated, 28 died (16 dying from lung cancer), and 73 were censored, with a mean overall survival time of 1,819.4 days (95% confidence interval [95% CI]: 1,481.2-2,157.5). One radiomic feature (the mean of the Fourier transform) presented a difference on Kaplan-Meier curves (p < 0.05). A high-risk group of patients was identified on the basis of high values for the mean of the Fourier transform. In that group, the mean survival time was 1,465.4 days (95% CI: 985.2-1,945.6), with a hazard ratio of 2.12 (95% CI: 1.01-4.48). We also identified a low-risk group, in which the mean of the Fourier transform was low (mean survival time of 2,164.8 days; 95% CI: 1,745.4-2,584.1). Conclusion A radiomic signature based on the Fourier transform correlates with overall survival, representing a prognostic biomarker for risk stratification in patients with lung cancer.
Collapse
Affiliation(s)
| | - Marcel Koenigkam-Santos
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Camila Vilas Boas Machado
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Matheus Calil Faleiros
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | | | | | - Alexandre Todorovic Fabro
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | | |
Collapse
|
10
|
Meyer HJ, Schneider I, Emmer A, Kornhuber M, Surov A. Associations between apparent diffusion coefficient values and histopathological tissue alterations in myopathies. Brain Behav 2020; 10:e01809. [PMID: 32860496 PMCID: PMC7667360 DOI: 10.1002/brb3.1809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/02/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Diffusion-weighted imaging (DWI) can reflect histopathologic changes in muscle disorders. The present study sought to elucidate possible associations between histopathology derived from muscle biopsies and DWI in myositis and other myopathies. METHODS Nineteen patients (10 women, 52.6%) with a mean age 51.43 ± 19 years were included in this retrospective study. Apparent diffusion coefficients (ADC) were evaluated with a histogram approach of the biopsied muscle. The histopathology analysis included the scoring systems proposed by Tateyama et al., Fanin et al., Allenbach et al. and immunhistochemical stainings for MHC, CD68, CD8, and CD4. RESULTS There was a tendency that skewness was lowered with increasing Tateyama score, but it did not reach statistical significance (p = .14). No statistical differences for the other scores were identified. There was a tendency that kurtosis was higher in MHC negative stained patient compared to positive patients, but statistically significance was not reached (p = .07). ADC histogram parameters did not correlate with CD68 and CD8 positive stained cells. There was a trend for skewness to correlate with the amount of CD4-positive cells (r = .57, p = .07). CONCLUSION The present study could not identify statistical significant associations between DWI and histopathology in muscle diseases based upon a small patient sample. Presumably, the investigated histopathology scores are more specific for certain disease aspects, whereas ADC values reflect the whole cellularity of the investigated muscle, which might cause the negative results.
Collapse
Affiliation(s)
- Hans-Jonas Meyer
- Department of Diagnostic and Interventional Radiology, University of Leipzig, Leipzig, Germany
| | - Ilka Schneider
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Alexander Emmer
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Malte Kornhuber
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Alexey Surov
- Department of Diagnostic and Interventional Radiology, University of Magdeburg, Magdeburg, Germany
| |
Collapse
|
11
|
An Automated Segmentation Pipeline for Intratumoural Regions in Animal Xenografts Using Machine Learning and Saturation Transfer MRI. Sci Rep 2020; 10:8063. [PMID: 32415137 PMCID: PMC7228927 DOI: 10.1038/s41598-020-64912-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/24/2020] [Indexed: 11/16/2022] Open
Abstract
Saturation transfer MRI can be useful in the characterization of different tumour types. It is sensitive to tumour metabolism, microstructure, and microenvironment. This study aimed to use saturation transfer to differentiate between intratumoural regions, demarcate tumour boundaries, and reduce data acquisition times by identifying the imaging scheme with the most impact on segmentation accuracy. Saturation transfer-weighted images were acquired over a wide range of saturation amplitudes and frequency offsets along with T1 and T2 maps for 34 tumour xenografts in mice. Independent component analysis and Gaussian mixture modelling were used to segment the images and identify intratumoural regions. Comparison between the segmented regions and histopathology indicated five distinct clusters: three corresponding to intratumoural regions (active tumour, necrosis/apoptosis, and blood/edema) and two extratumoural (muscle and a mix of muscle and connective tissue). The fraction of tumour voxels segmented as necrosis/apoptosis quantitatively matched those calculated from TUNEL histopathological assays. An optimal protocol was identified providing reasonable qualitative agreement between MRI and histopathology and consisting of T1 and T2 maps and 22 magnetization transfer (MT)-weighted images. A three-image subset was identified that resulted in a greater than 90% match in positive and negative predictive value of tumour voxels compared to those found using the entire 24-image dataset. The proposed algorithm can potentially be used to develop a robust intratumoural segmentation method.
Collapse
|
12
|
Olin A, Krogager L, Rasmussen JH, Andersen FL, Specht L, Beyer T, Kjaer A, Fischer BM, Hansen AE. Preparing data for multiparametric PET/MR imaging: Influence of PET point spread function modelling and EPI distortion correction on the spatial correlation of [18F]FDG-PET and diffusion-weighted MRI in head and neck cancer. Phys Med 2019; 61:1-7. [DOI: 10.1016/j.ejmp.2019.04.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/18/2019] [Accepted: 04/08/2019] [Indexed: 10/27/2022] Open
|
13
|
Kim J, Ryu SY, Lee SH, Lee HY, Park H. Clustering approach to identify intratumour heterogeneity combining FDG PET and diffusion-weighted MRI in lung adenocarcinoma. Eur Radiol 2019; 29:468-475. [PMID: 29922931 DOI: 10.1007/s00330-018-5590-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/13/2018] [Accepted: 06/04/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Malignant tumours consist of biologically heterogeneous components; identifying and stratifying those various subregions is an important research topic. We aimed to show the effectiveness of an intratumour partitioning method using clustering to identify highly aggressive tumour subregions, determining prognosis based on pre-treatment PET and DWI in stage IV lung adenocarcinoma. METHODS Eighteen patients who underwent both baseline PET and DWI were recruited. Pre-treatment imaging of SUV and ADC values were used to form intensity vectors within manually specified ROIs. We applied k-means clustering to intensity vectors to yield distinct subregions, then chose the subregion that best matched the criteria for high SUV and low ADC to identify tumour subregions with high aggressiveness. We stratified patients into high- and low-risk groups based on subregion volume with high aggressiveness and conducted survival analyses. This approach is referred to as the partitioning approach. For comparison, we computed tumour subregions with high aggressiveness without clustering and repeated the described procedure; this is referred to as the voxel-wise approach. RESULTS The partitioning approach led to high-risk (median SUVmax = 14.25 and median ADC = 1.26x10-3 mm2/s) and low-risk (median SUVmax = 14.64 and median ADC = 1.09x10-3 mm2/s) subgroups. Our partitioning approach identified significant differences in survival between high- and low-risk subgroups (hazard ratio, 4.062, 95% confidence interval, 1.21 - 13.58, p-value: 0.035). The voxel-wise approach did not identify significant differences in survival between high- and low-risk subgroups (p-value: 0.325). CONCLUSION Our partitioning approach identified intratumour subregions that were predictors of survival. KEY POINTS • Multimodal imaging of PET and DWI is useful for assessing intratumour heterogeneity. • Data-driven clustering identified subregions which might be highly aggressive for lung adenocarcinoma. • The data-driven partitioning results might be predictors of survival.
Collapse
Affiliation(s)
- Jonghoon Kim
- Department of Electronic Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Korea
| | - Seong-Yoon Ryu
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Ilwon-Dong, Kangnam-Ku, Seoul, 06315, Korea
| | - Seung-Hak Lee
- Department of Electronic Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Korea
| | - Ho Yun Lee
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Ilwon-Dong, Kangnam-Ku, Seoul, 06315, Korea.
| | - Hyunjin Park
- School of Electronic and Electrical Engineering, Sungkyunkwan University, Suwon, 16419, Korea.
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea.
| |
Collapse
|
14
|
Xing S, Freeman CR, Jung S, Turcotte R, Levesque IR. Probabilistic classification of tumour habitats in soft tissue sarcoma. NMR IN BIOMEDICINE 2018; 31:e4000. [PMID: 30113738 DOI: 10.1002/nbm.4000] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/27/2018] [Accepted: 07/02/2018] [Indexed: 06/08/2023]
Abstract
The purpose of this work is to propose a method to characterize tumour heterogeneity on MRI, using probabilistic classification based on a reference tissue. The method uses maps of the apparent diffusion coefficient (ADC), T2 relaxation, and a calculated map representing high-b-value diffusion-weighted MRI (denoted simDWI) to identify up to five habitats (i.e. sub-regions) of tumours. In this classification method, the parameter values (ADC, T2 , and simDWI) from each tumour voxel are compared against the corresponding parameter probability distributions in a reference tissue. The probability that a tumour voxel belongs to a specific habitat is the joint probability for all parameters. The classification can be visualized using a custom colour scheme. The proposed method was applied to data from seven patients with biopsy-confirmed soft tissue sarcoma, at three time-points over the course of pre-operative radiotherapy. Fast-spin-echo images with two different echo times and diffusion MRI with three b-values were obtained and used as inputs to the method. Imaging findings were compared with pathology reports from pre-radiotherapy biopsy and post-surgical resection. Regions of hypercellularity, high-T2 proteinaceous fluid, necrosis, collagenous stroma, and fibrosis were identified within soft tissue sarcoma. The classifications were qualitatively consistent with pathological observations. The percentage of necrosis on imaging correlated strongly with necrosis estimated from FDG-PET before radiotherapy (R2 = 0.97) and after radiotherapy (R2 = 0.96). The probabilistic classification method identifies realistic habitats and reflects the complex microenvironment of tumours, as demonstrated in soft tissue sarcoma.
Collapse
Affiliation(s)
- Shu Xing
- Medical Physics Unit, McGill University, Montreal, Canada
- Department of Physics, McGill University, Montreal, Canada
| | - Carolyn R Freeman
- Radiation Oncology, McGill University Health Centre, Montreal, Canada
| | - Sungmi Jung
- Department of Pathology, McGill University Health Centre, Montreal, Canada
| | - Robert Turcotte
- Department of Surgery, McGill University Health Centre, Montreal, Canada
| | - Ives R Levesque
- Medical Physics Unit, McGill University, Montreal, Canada
- Department of Physics, McGill University, Montreal, Canada
- Research Institute of the McGill University Health Centre, Montreal, Canada
| |
Collapse
|
15
|
Hu T, Wang S, Huang L, Wang J, Shi D, Li Y, Tong T, Peng W. A clinical-radiomics nomogram for the preoperative prediction of lung metastasis in colorectal cancer patients with indeterminate pulmonary nodules. Eur Radiol 2018; 29:439-449. [DOI: 10.1007/s00330-018-5539-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 05/01/2018] [Accepted: 05/14/2018] [Indexed: 12/19/2022]
|
16
|
Mannheim JG, Schmid AM, Schwenck J, Katiyar P, Herfert K, Pichler BJ, Disselhorst JA. PET/MRI Hybrid Systems. Semin Nucl Med 2018; 48:332-347. [PMID: 29852943 DOI: 10.1053/j.semnuclmed.2018.02.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Over the last decade, the combination of PET and MRI in one system has proven to be highly successful in basic preclinical research, as well as in clinical research. Nowadays, PET/MRI systems are well established in preclinical imaging and are progressing into clinical applications to provide further insights into specific diseases, therapeutic assessments, and biological pathways. Certain challenges in terms of hardware had to be resolved concurrently with the development of new techniques to be able to reach the full potential of both combined techniques. This review provides an overview of these challenges and describes the opportunities that simultaneous PET/MRI systems can exploit in comparison with stand-alone or other combined hybrid systems. New approaches were developed for simultaneous PET/MRI systems to correct for attenuation of 511 keV photons because MRI does not provide direct information on gamma photon attenuation properties. Furthermore, new algorithms to correct for motion were developed, because MRI can accurately detect motion with high temporal resolution. The additional information gained by the MRI can be employed to correct for partial volume effects as well. The development of new detector designs in combination with fast-decaying scintillator crystal materials enabled time-of-flight detection and incorporation in the reconstruction algorithms. Furthermore, this review lists the currently commercially available systems both for preclinical and clinical imaging and provides an overview of applications in both fields. In this regard, special emphasis has been placed on data analysis and the potential for both modalities to evolve with advanced image analysis tools, such as cluster analysis and machine learning.
Collapse
Affiliation(s)
- Julia G Mannheim
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Andreas M Schmid
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Johannes Schwenck
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany; Department of Nuclear Medicine and Clinical Molecular Imaging, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Prateek Katiyar
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Kristina Herfert
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Bernd J Pichler
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany.
| | - Jonathan A Disselhorst
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| |
Collapse
|
17
|
Abstract
Imaging provides an insight into biological patho-mechanisms of diseases. However, the link between the imaging phenotype and the underlying molecular processes is often not well understood. Methods such as metabolomics and proteomics reveal detailed information about these processes. Unfortunately, they provide no spatial information and thus cannot be easily correlated with functional imaging. We have developed an image-guided milling machine and unique workflows to precisely isolate tissue samples based on imaging data. The tissue samples remain cooled during the entire procedure, preventing sample degradation. This enables us to correlate, at an unprecedented spatial precision, comprehensive imaging information with metabolomics and proteomics data, leading to a better understanding of diseases. Phenotypic heterogeneity is commonly observed in diseased tissue, specifically in tumors. Multimodal imaging technologies can reveal tissue heterogeneity noninvasively in vivo, enabling imaging-based profiling of receptors, metabolism, morphology, or function on a macroscopic scale. In contrast, in vitro multiomics, immunohistochemistry, or histology techniques accurately characterize these heterogeneities in the cellular and subcellular scales in a more comprehensive but ex vivo manner. The complementary in vivo and ex vivo information would provide an enormous potential to better characterize a disease. However, this requires spatially accurate coregistration of these data by image-driven sampling as well as fast sample-preparation methods. Here, a unique image-guided milling machine and workflow for precise extraction of tissue samples from small laboratory animals or excised organs has been developed and evaluated. The samples can be delineated on tomographic images as volumes of interest and can be extracted with a spatial accuracy better than 0.25 mm. The samples remain cooled throughout the procedure to ensure metabolic stability, a precondition for accurate in vitro analysis.
Collapse
|
18
|
Mancini M, Summers P, Faita F, Brunetto MR, Callea F, De Nicola A, Di Lascio N, Farinati F, Gastaldelli A, Gridelli B, Mirabelli P, Neri E, Salvadori PA, Rebelos E, Tiribelli C, Valenti L, Salvatore M, Bonino F. Digital liver biopsy: Bio-imaging of fatty liver for translational and clinical research. World J Hepatol 2018; 10:231-245. [PMID: 29527259 PMCID: PMC5838442 DOI: 10.4254/wjh.v10.i2.231] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 01/27/2018] [Accepted: 02/25/2018] [Indexed: 02/06/2023] Open
Abstract
The rapidly growing field of functional, molecular and structural bio-imaging is providing an extraordinary new opportunity to overcome the limits of invasive liver biopsy and introduce a "digital biopsy" for in vivo study of liver pathophysiology. To foster the application of bio-imaging in clinical and translational research, there is a need to standardize the methods of both acquisition and the storage of the bio-images of the liver. It can be hoped that the combination of digital, liquid and histologic liver biopsies will provide an innovative synergistic tri-dimensional approach to identifying new aetiologies, diagnostic and prognostic biomarkers and therapeutic targets for the optimization of personalized therapy of liver diseases and liver cancer. A group of experts of different disciplines (Special Interest Group for Personalized Hepatology of the Italian Association for the Study of the Liver, Institute for Biostructures and Bio-imaging of the National Research Council and Bio-banking and Biomolecular Resources Research Infrastructure) discussed criteria, methods and guidelines for facilitating the requisite application of data collection. This manuscript provides a multi-Author review of the issue with special focus on fatty liver.
Collapse
Affiliation(s)
- Marcello Mancini
- Institute of Biostructure and Bioimaging, National Research Council, Naples 80145, Italy
| | - Paul Summers
- European Institute of Oncology (IEO) IRCCS, Milan 20141, Italy
| | - Francesco Faita
- Institute of Clinical Physiology (IFC), National Research Council (CNR), Pisa 56124, Italy
| | - Maurizia R Brunetto
- Hepatology Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa 56125, Italy
| | - Francesco Callea
- Department of Pathology, Children Hospital Bambino Gesù IRCCS, Rome 00165, Italy
| | | | - Nicole Di Lascio
- Institute of Clinical Physiology (IFC), National Research Council (CNR), Pisa 56124, Italy
| | - Fabio Farinati
- Department of Gastroenterology, Oncology and Surgical Sciences, University of Padua, Padua 35121, Italy
| | - Amalia Gastaldelli
- Cardio-metabolic Risk Laboratory, Institute of Clinical Physiology (IFC), National Research Council (CNR), Pisa 56124, Italy
| | - Bruno Gridelli
- Institute for Health, University of Pittsburgh Medical Center (UPMC), Chianciano Terme 53042, Italy
| | | | - Emanuele Neri
- Diagnostic Radiology 3, Department of Translational Research, University of Pisa and "Ospedale S. Chiara" AOUP, Pisa 56126, Italy
| | - Piero A Salvadori
- Institute of Clinical Physiology (IFC), National Research Council (CNR), Pisa 56124, Italy
| | - Eleni Rebelos
- Hepatology Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa 56125, Italy
| | - Claudio Tiribelli
- Fondazione Italiana Fegato (FIF), Area Science Park, Campus Basovizza, Trieste 34012, Italy
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano and Department of Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milan 20122, Italy
| | | | - Ferruccio Bonino
- Institute of Biostructure and Bioimaging, National Research Council, Naples 80145, Italy
| |
Collapse
|
19
|
Katiyar P, Divine MR, Kohlhofer U, Quintanilla-Martinez L, Schölkopf B, Pichler BJ, Disselhorst JA. A Novel Unsupervised Segmentation Approach Quantifies Tumor Tissue Populations Using Multiparametric MRI: First Results with Histological Validation. Mol Imaging Biol 2018; 19:391-397. [PMID: 27734253 PMCID: PMC5332060 DOI: 10.1007/s11307-016-1009-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Purpose We aimed to precisely estimate intra-tumoral heterogeneity using spatially regularized spectral clustering (SRSC) on multiparametric MRI data and compare the efficacy of SRSC with the previously reported segmentation techniques in MRI studies. Procedures Six NMRI nu/nu mice bearing subcutaneous human glioblastoma U87 MG tumors were scanned using a dedicated small animal 7T magnetic resonance imaging (MRI) scanner. The data consisted of T2 weighted images, apparent diffusion coefficient maps, and pre- and post-contrast T2 and T2* maps. Following each scan, the tumors were excised into 2–3-mm thin slices parallel to the axial field of view and processed for histological staining. The MRI data were segmented using SRSC, K-means, fuzzy C-means, and Gaussian mixture modeling to estimate the fractional population of necrotic, peri-necrotic, and viable regions and validated with the fractional population obtained from histology. Results While the aforementioned methods overestimated peri-necrotic and underestimated viable fractions, SRSC accurately predicted the fractional population of all three tumor tissue types and exhibited strong correlations (rnecrotic = 0.92, rperi-necrotic = 0.82 and rviable = 0.98) with the histology. Conclusions The precise identification of necrotic, peri-necrotic and viable areas using SRSC may greatly assist in cancer treatment planning and add a new dimension to MRI-guided tumor biopsy procedures. Electronic supplementary material The online version of this article (doi:10.1007/s11307-016-1009-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Prateek Katiyar
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Roentgenweg 13, 72076, Tuebingen, Germany.
- Max Planck Institute for Intelligent Systems, Tuebingen, Germany.
| | - Mathew R Divine
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Roentgenweg 13, 72076, Tuebingen, Germany
| | - Ursula Kohlhofer
- Institute of Pathology and Neuropathology, Eberhard Karls University Tuebingen and Comprehensive Cancer Center, University Hospital Tuebingen, Tuebingen, Germany
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, Eberhard Karls University Tuebingen and Comprehensive Cancer Center, University Hospital Tuebingen, Tuebingen, Germany
| | | | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Roentgenweg 13, 72076, Tuebingen, Germany
| | - Jonathan A Disselhorst
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Roentgenweg 13, 72076, Tuebingen, Germany
| |
Collapse
|
20
|
Lee G, Bak SH, Lee HY. CT Radiomics in Thoracic Oncology: Technique and Clinical Applications. Nucl Med Mol Imaging 2017; 52:91-98. [PMID: 29662557 DOI: 10.1007/s13139-017-0506-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/02/2017] [Accepted: 11/16/2017] [Indexed: 11/26/2022] Open
Abstract
Precision medicine offers better treatment options and improved survival for cancer patients based on individual variability. As the success of precision medicine depends on robust biomarkers, the requirement for improved imaging biomarkers that reflect tumor biology has grown exponentially. Radiomics, the field of study in which high-throughput data are generated and large amounts of advanced quantitative features are extracted from medical images, has shown great potential as a source of quantitative biomarkers in the field of oncology. Radiomics provides quantitative information about the morphology, texture, and intratumoral heterogeneity of the tumor itself as well as features related to pulmonary function. Hence, radiomics data can be used to build descriptive and predictive clinical models that relate imaging characteristics to tumor biology phenotypes. In this review, we describe the workflow of CT radiomics, types of CT radiomics, and its clinical application in thoracic oncology.
Collapse
Affiliation(s)
- Geewon Lee
- 1Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-Ro, Gangnam-gu, Seoul, 06351 South Korea
- Department of Radiology and Medical Research Institute, Pusan National University Hospital, Pusan National University School of Medicine, Busan, South Korea
| | - So Hyeon Bak
- 1Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-Ro, Gangnam-gu, Seoul, 06351 South Korea
- 3Department of Radiology, Kangwon National University Hospital, Chuncheon, South Korea
| | - Ho Yun Lee
- 1Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-Ro, Gangnam-gu, Seoul, 06351 South Korea
| |
Collapse
|
21
|
Zhu T, Das S, Wong TZ. Integration of PET/MR Hybrid Imaging into Radiation Therapy Treatment. Magn Reson Imaging Clin N Am 2017; 25:377-430. [PMID: 28390536 DOI: 10.1016/j.mric.2017.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Hybrid PET/MR imaging is in early development for treatment planning. This article briefly reviews research and clinical applications of PET/MR imaging in radiation oncology. With improvements in workflow, more specific tracers, and fast and robust acquisition protocols, PET/MR imaging will play an increasingly important role in better target delineation for treatment planning and have clear advantages in the evaluation of tumor response and in a better understanding of tumor heterogeneity. With advances in treatment delivery and the potential of integrating PET/MR imaging with research on radiomics for radiation oncology, quantitative and physiologic information could lead to more precise and personalized RT.
Collapse
Affiliation(s)
- Tong Zhu
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, 101 Manning Drive, Chapel Hill, NC 27599, USA
| | - Shiva Das
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, 101 Manning Drive, Chapel Hill, NC 27599, USA
| | - Terence Z Wong
- Department of Radiology, University of North Carolina at Chapel Hill, 101 Manning Drive, Chapel Hill, NC 27599, USA.
| |
Collapse
|
22
|
Lee G, Lee HY, Ko ES, Jeong WK. Radiomics and imaging genomics in precision medicine. PRECISION AND FUTURE MEDICINE 2017. [DOI: 10.23838/pfm.2017.00101] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
23
|
Pant K, Sedláček O, Nadar RA, Hrubý M, Stephan H. Radiolabelled Polymeric Materials for Imaging and Treatment of Cancer: Quo Vadis? Adv Healthc Mater 2017; 6. [PMID: 28218487 DOI: 10.1002/adhm.201601115] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/24/2016] [Indexed: 12/15/2022]
Abstract
Owing to their tunable blood circulation time and suitable plasma stability, polymer-based nanomaterials hold a great potential for designing and utilising multifunctional nanocarriers for efficient imaging and effective treatment of cancer. When tagged with appropriate radionuclides, they may allow for specific detection (diagnosis) as well as the destruction of tumours (therapy) or even customization of materials, aiming to both diagnosis and therapy (theranostic approach). This review provides an overview of recent developments of radiolabelled polymeric nanomaterials (natural and synthetic polymers) for molecular imaging of cancer, specifically, applying nuclear techniques such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT). Different approaches to radiolabel polymers are evaluated from the methodical radiochemical point of view. This includes new bifunctional chelating agents (BFCAs) for radiometals as well as novel labelling methods. Special emphasis is given to eligible strategies employed to evade the mononuclear phagocytic system (MPS) in view of efficient targeting. The discussion encompasses promising strategies currently employed as well as emerging possibilities in radionuclide-based cancer therapy. Key issues involved in the clinical translation of radiolabelled polymers and future scopes of this intriguing research field are also discussed.
Collapse
Affiliation(s)
- Kritee Pant
- Helmholtz-Zentrum Dresden-Rossendorf; Institute of Radiopharmaceutical Cancer Research; Bautzner Landstraße 400 01328 Dresden Germany
| | - Ondřej Sedláček
- Institute of Macromolecular Chemistry; The Academy of Sciences of the Czech Republic; Heyrovského námeˇstí 2 16206 Prague 6 Czech Republic
| | - Robin A. Nadar
- Helmholtz-Zentrum Dresden-Rossendorf; Institute of Radiopharmaceutical Cancer Research; Bautzner Landstraße 400 01328 Dresden Germany
| | - Martin Hrubý
- Institute of Macromolecular Chemistry; The Academy of Sciences of the Czech Republic; Heyrovského námeˇstí 2 16206 Prague 6 Czech Republic
| | - Holger Stephan
- Helmholtz-Zentrum Dresden-Rossendorf; Institute of Radiopharmaceutical Cancer Research; Bautzner Landstraße 400 01328 Dresden Germany
| |
Collapse
|
24
|
Katiyar P, Divine MR, Kohlhofer U, Quintanilla-Martinez L, Schölkopf B, Pichler BJ, Disselhorst JA. Spectral Clustering Predicts Tumor Tissue Heterogeneity Using Dynamic 18F-FDG PET: A Complement to the Standard Compartmental Modeling Approach. J Nucl Med 2016; 58:651-657. [PMID: 27811120 DOI: 10.2967/jnumed.116.181370] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/19/2016] [Indexed: 12/11/2022] Open
Abstract
In this study, we described and validated an unsupervised segmentation algorithm for the assessment of tumor heterogeneity using dynamic 18F-FDG PET. The aim of our study was to objectively evaluate the proposed method and make comparisons with compartmental modeling parametric maps and SUV segmentations using simulations of clinically relevant tumor tissue types. Methods: An irreversible 2-tissue-compartmental model was implemented to simulate clinical and preclinical 18F-FDG PET time-activity curves using population-based arterial input functions (80 clinical and 12 preclinical) and the kinetic parameter values of 3 tumor tissue types. The simulated time-activity curves were corrupted with different levels of noise and used to calculate the tissue-type misclassification errors of spectral clustering (SC), parametric maps, and SUV segmentation. The utility of the inverse noise variance- and Laplacian score-derived frame weighting schemes before SC was also investigated. Finally, the SC scheme with the best results was tested on a dynamic 18F-FDG measurement of a mouse bearing subcutaneous colon cancer and validated using histology. Results: In the preclinical setup, the inverse noise variance-weighted SC exhibited the lowest misclassification errors (8.09%-28.53%) at all noise levels in contrast to the Laplacian score-weighted SC (16.12%-31.23%), unweighted SC (25.73%-40.03%), parametric maps (28.02%-61.45%), and SUV (45.49%-45.63%) segmentation. The classification efficacy of both weighted SC schemes in the clinical case was comparable to the unweighted SC. When applied to the dynamic 18F-FDG measurement of colon cancer, the proposed algorithm accurately identified densely vascularized regions from the rest of the tumor. In addition, the segmented regions and clusterwise average time-activity curves showed excellent correlation with the tumor histology. Conclusion: The promising results of SC mark its position as a robust tool for quantification of tumor heterogeneity using dynamic PET studies. Because SC tumor segmentation is based on the intrinsic structure of the underlying data, it can be easily applied to other cancer types as well.
Collapse
Affiliation(s)
- Prateek Katiyar
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany.,Max Planck Institute for Intelligent Systems, Tuebingen, Germany; and
| | - Mathew R Divine
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Ursula Kohlhofer
- Institute of Pathology and Neuropathology, Eberhard Karls University Tuebingen and Comprehensive Cancer Center, University Hospital Tuebingen, Tuebingen, Germany
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, Eberhard Karls University Tuebingen and Comprehensive Cancer Center, University Hospital Tuebingen, Tuebingen, Germany
| | | | - Bernd J Pichler
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Jonathan A Disselhorst
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| |
Collapse
|
25
|
Timm KN, Kennedy BWC, Brindle KM. Imaging Tumor Metabolism to Assess Disease Progression and Treatment Response. Clin Cancer Res 2016; 22:5196-5203. [PMID: 27609841 PMCID: PMC5321522 DOI: 10.1158/1078-0432.ccr-16-0159] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 08/09/2016] [Indexed: 12/26/2022]
Abstract
Changes in tumor metabolism may accompany disease progression and can occur following treatment, often before there are changes in tumor size. We focus here on imaging methods that can be used to image various aspects of tumor metabolism, with an emphasis on methods that can be used for tumor grading, assessing disease progression, and monitoring treatment response. Clin Cancer Res; 22(21); 5196-203. ©2016 AACR.
Collapse
Affiliation(s)
- Kerstin N Timm
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Brett W C Kennedy
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Kevin M Brindle
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
26
|
Radiomics and its emerging role in lung cancer research, imaging biomarkers and clinical management: State of the art. Eur J Radiol 2016; 86:297-307. [PMID: 27638103 DOI: 10.1016/j.ejrad.2016.09.005] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 09/09/2016] [Indexed: 12/29/2022]
Abstract
With the development of functional imaging modalities we now have the ability to study the microenvironment of lung cancer and its genomic instability. Radiomics is defined as the use of automated or semi-automated post-processing and analysis of large amounts of quantitative imaging features that can be derived from medical images. The automated generation of these analytical features helps to quantify a number of variables in the imaging assessment of lung malignancy. These imaging features include: tumor spatial complexity, elucidation of the tumor genomic heterogeneity and composition, subregional identification in terms of tumor viability or aggressiveness, and response to chemotherapy and/or radiation. Therefore, a radiomic approach can help to reveal unique information about tumor behavior. Currently available radiomic features can be divided into four major classes: (a) morphological, (b) statistical, (c) regional, and (d) model-based. Each category yields quantitative parameters that reflect specific aspects of a tumor. The major challenge is to integrate radiomic data with clinical, pathological, and genomic information to decode the different types of tissue biology. There are many currently available radiomic studies on lung cancer for which there is a need to summarize the current state of the art.
Collapse
|