1
|
Nabar SJ, Shet T, Malpani B, Hassan P, Basu S. Technetium-99m-labeled stealth liposomes: A new Strategy to Identify metastasis in tumour model. Appl Radiat Isot 2025; 218:111709. [PMID: 39923339 DOI: 10.1016/j.apradiso.2025.111709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/11/2025]
Abstract
OBJECTIVE Stealth liposomes are useful carriers for delivering drugs to cancer sites. In this paper we describe the preparation and evaluation of 99mTc labelled stealth liposomes (PEG-Liposomes) as potential radiopharmaceuticals for SPECT imaging of cancer. This study is first to describe targeted localisation of radiolabeled stealth liposomes in tumourous cells, sparing normal cells of various organs in metastatic L1210 mouse tumour model in BDF1 mice. METHODS Glutathione encapsulated stealth liposomes were made using lipid film hydration technique followed by probe sonication. Liposomes were radiolabeled using a lipophilic technetium complex, 99mTc-d,l-HMPAO. Labelled liposomes were purified by gel chromatography over sephadex G25 column. The tumour model was developed in immuno-competent BDF1 mice (F1DBA/2/C57-BL6 crosses) by injecting L1210 cells intraperitoneally. Biodistribution studies of the 99mTc- stealth liposomes were done in both tumour induced and normal mice. Histopathologic studies were done by excising the organs and the radioactivity in various sections was detected by autoradiography. RESULTS The stealth liposomes could be synthesized as per a reported procedure and it showed similar retention factor in thin layer chromatography. Liposomes could be radiolabelled by using 99mTc-d,lHMPAO. Purification over sephadex G 25 column yielded radioachemical purity greater than 95%. Biodistribution studies and autoradiography studies showed significantly higher accumulation of 99mTc labelled stealth liposomes in liver, pancreas and ascitic fluid of the tumour induced mice as compared to normal mice. CONCLUSIONS 99mTc labelled stealth liposomes having radiochemical purity greater than 95% could be prepared which showed higher uptake in tumour.
Collapse
Affiliation(s)
- Swapna J Nabar
- Homi Bhabha National Institute and Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Campus, Annex Building, JerbaiWadia Road, Dadar East, Mumbai, 400012, India.
| | - Tanuja Shet
- Department of Pathology, Tata Memorial Hospital, Parel, Mumbai, 400012, India
| | - Basant Malpani
- Homi Bhabha National Institute and Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Campus, Annex Building, JerbaiWadia Road, Dadar East, Mumbai, 400012, India
| | - Puthusserickal Hassan
- Homi Bhabha National Institute, Bhabha Atomic Research Centre, Trombay, Mumbai, 400012, India
| | - Sandip Basu
- Homi Bhabha National Institute and Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Campus, Annex Building, JerbaiWadia Road, Dadar East, Mumbai, 400012, India
| |
Collapse
|
2
|
Smeets EMM, Trajkovic-Arsic M, Geijs D, Karakaya S, van Zanten M, Brosens LAA, Feuerecker B, Gotthardt M, Siveke JT, Braren R, Ciompi F, Aarntzen EHJG. Histology-Based Radiomics for [ 18F]FDG PET Identifies Tissue Heterogeneity in Pancreatic Cancer. J Nucl Med 2024; 65:1151-1159. [PMID: 38782455 DOI: 10.2967/jnumed.123.266262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 04/22/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
Radiomics features can reveal hidden patterns in a tumor but usually lack an underlying biologic rationale. In this work, we aimed to investigate whether there is a correlation between radiomics features extracted from [18F]FDG PET images and histologic expression patterns of a glycolytic marker, monocarboxylate transporter-4 (MCT4), in pancreatic cancer. Methods: A cohort of pancreatic ductal adenocarcinoma patients (n = 29) for whom both tumor cross sections and [18F]FDG PET/CT scans were available was used to develop an [18F]FDG PET radiomics signature. By using immunohistochemistry for MCT4, we computed density maps of MCT4 expression and extracted pathomics features. Cluster analysis identified 2 subgroups with distinct MCT4 expression patterns. From corresponding [18F]FDG PET scans, radiomics features that associate with the predefined MCT4 subgroups were identified. Results: Complex heat map visualization showed that the MCT4-high/heterogeneous subgroup was correlating with a higher MCT4 expression level and local variation. This pattern linked to a specific [18F]FDG PET signature, characterized by a higher SUVmean and SUVmax and second-order radiomics features, correlating with local variation. This MCT4-based [18F]FDG PET signature of 7 radiomics features demonstrated prognostic value in an independent cohort of pancreatic cancer patients (n = 71) and identified patients with worse survival. Conclusion: Our cross-modal pipeline allows the development of PET scan signatures based on immunohistochemical analysis of markers of a particular biologic feature, here demonstrated on pancreatic cancer using intratumoral MCT4 expression levels to select [18F]FDG PET radiomics features. This study demonstrated the potential of radiomics scores to noninvasively capture intratumoral marker heterogeneity and identify a subset of pancreatic ductal adenocarcinoma patients with a poor prognosis.
Collapse
Affiliation(s)
- Esther M M Smeets
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marija Trajkovic-Arsic
- German Cancer Consortium, partner site Essen, a partnership between DKFZ and University Hospital Essen, Essen, Germany
- Bridge Institute of Experimental Tumor Therapy and Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Daan Geijs
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sinan Karakaya
- German Cancer Consortium, partner site Essen, a partnership between DKFZ and University Hospital Essen, Essen, Germany
- Bridge Institute of Experimental Tumor Therapy and Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Monica van Zanten
- Department of Pathology, Canisius Wilhelmina Ziekenhuis, Nijmegen, The Netherlands
| | - Lodewijk A A Brosens
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Benedikt Feuerecker
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
- Department of Radiology, School of Medicine, Technical University of Munich, Munich, Germany
- German Cancer Consortium, partner site Munich, a partnership between DKFZ and Technical University of Munich, Munich, Germany
- Department of Radiology, Ludwig Maximilians University, Munich, Germany; and
| | - Martin Gotthardt
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jens T Siveke
- German Cancer Consortium, partner site Essen, a partnership between DKFZ and University Hospital Essen, Essen, Germany
- Bridge Institute of Experimental Tumor Therapy and Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- National Center for Tumor Diseases West, Campus Essen, Essen, Germany
| | - Rickmer Braren
- Department of Radiology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Francesco Ciompi
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Erik H J G Aarntzen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands;
| |
Collapse
|
3
|
Naydenov N, Teplov A, Zirakchian MZ, Ruan S, Chu BP, Serencsits B, Iraca M, Talarico O, Miller B, Kunin H, Schwartz J, Kesner A, Furenlid LR, Dauer L, Yagi Y, Humm JL, Zanzonico P, Sofocleous CT, Kirov AS. Yttrium-90 Activity Quantification in PET/CT-Guided Biopsy Specimens from Colorectal Hepatic Metastases Immediately after Transarterial Radioembolization Using Micro-CT and Autoradiography. J Vasc Interv Radiol 2023; 34:1556-1564.e4. [PMID: 37201655 PMCID: PMC11163896 DOI: 10.1016/j.jvir.2023.05.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 04/13/2023] [Accepted: 05/08/2023] [Indexed: 05/20/2023] Open
Abstract
PURPOSE To evaluate the yttrium-90 (90Y) activity distribution in biopsy tissue samples of the treated liver to quantify the dose with higher spatial resolution than positron emission tomography (PET) for accurate investigation of correlations with microscopic biological effects and to evaluate the radiation safety of this procedure. MATERIALS AND METHODS Eighty-six core biopsy specimens were obtained from 18 colorectal liver metastases (CLMs) immediately after 90Y transarterial radioembolization (TARE) with either resin or glass microspheres using real-time 90Y PET/CT guidance in 17 patients. A high-resolution micro-computed tomography (micro-CT) scanner was used to image the microspheres in part of the specimens and allow quantification of 90Y activity directly or by calibrating autoradiography (ARG) images. The mean doses to the specimens were derived from the measured specimens' activity concentrations and from the PET/CT scan at the location of the biopsy needle tip for all cases. Staff exposures were monitored. RESULTS The mean measured 90Y activity concentration in the CLM specimens at time of infusion was 2.4 ± 4.0 MBq/mL. The biopsies revealed higher activity heterogeneity than PET. Radiation exposure to the interventional radiologists during post-TARE biopsy procedures was minimal. CONCLUSIONS Counting the microspheres and measuring the activity in biopsy specimens obtained after TARE are safe and feasible and can be used to determine the administered activity and its distribution in the treated and biopsied liver tissue with high spatial resolution. Complementing 90Y PET/CT imaging with this approach promises to yield more accurate direct correlation of histopathological changes and absorbed dose in the examined specimens.
Collapse
Affiliation(s)
- Nicola Naydenov
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexei Teplov
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Shutian Ruan
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bae P Chu
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brian Serencsits
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marisa Iraca
- University of Rhode Island, Kingston, Rhode Island
| | - Olga Talarico
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Henry Kunin
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jazmin Schwartz
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Adam Kesner
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Larry Dauer
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yukako Yagi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - John L Humm
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pat Zanzonico
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Assen S Kirov
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
4
|
Dzaye O, Cornelis FH, Kunin HS, Sofocleous CT. Advancements and Future Outlook of PET/CT-Guided Interventions. Tech Vasc Interv Radiol 2023; 26:100916. [PMID: 38071029 DOI: 10.1016/j.tvir.2023.100916] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Advancements in minimally invasive technology, coupled with imaging breakthroughs, have empowered the field of interventional radiology to achieve unparalleled precision in image-guided diagnosis and treatment while simultaneously reducing periprocedural morbidity. Molecular imaging, which provides valuable physiological and metabolic information alongside anatomical localization, can expand the capabilities of image-guided interventions. Among various molecular imaging techniques, positron emission tomography (PET) stands out for its superior spatial resolution and ability to acquire quantitative data. PET has emerged as a crucial tool for oncologic imaging and plays a pivotal role in both staging and the assessment of treatment responses. Typically used in combination with computed tomography (CT) (PET/CT) and occasionally with magnetic resonance imaging MRI (PET/MRI), PET as a hybrid imaging approach offers enhanced insights into disease progression and response. In recent years, PET has also found its way into image-guided interventions, especially within the rapidly expanding field of interventional oncology. This review aims to explore the current and evolving role of metabolic imaging, specifically PET, in interventional oncology. By delving into the unique advantages and applications of PET in guiding oncological interventions and assessing response, we seek to highlight the increasing significance of this modality in the realm of interventional radiology.
Collapse
Affiliation(s)
- Omar Dzaye
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY; Molecular Imaging & Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Francois H Cornelis
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Henry S Kunin
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Constantinos T Sofocleous
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY.
| |
Collapse
|
5
|
Debacker JM, Maris L, Cordier F, Creytens D, Deron P, Descamps B, D'Asseler Y, De Man K, Keereman V, Libbrecht S, Schelfhout V, Van de Vijver K, Vanhove C, Huvenne W. Direct co-registration of [ 18F]FDG uptake and histopathology in surgically excised malignancies of the head and neck: a feasibility study. Eur J Nucl Med Mol Imaging 2023; 50:2127-2139. [PMID: 36854863 DOI: 10.1007/s00259-023-06153-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 02/13/2023] [Indexed: 03/02/2023]
Abstract
PURPOSE Recent technical advancements in PET imaging have improved sensitivity and spatial resolution. Consequently, clinical nuclear medicine will be confronted with PET images on a previously unfamiliar resolution. To better understand [18F]FDG distribution at submillimetric scale, a direct correlation of radionuclide-imaging and histopathology is required. METHODS A total of five patients diagnosed with a malignancy of the head and neck were injected with a clinical activity of [18F]FDG before undergoing surgical resection. The resected specimen was imaged using a preclinical high-resolution PET/CT, followed by slicing of the specimen. Multiple slices were rescanned using a micro-PET/CT device, and one of the slices was snap-frozen for frozen sections. Frozen sections were placed on an autoradiographic film, followed by haematoxylin and eosin staining to prepare them for histopathological assessment. The results from both autoradiography and histopathology were co-registered using an iterative co-registration algorithm, and regions of interest were identified to study radiotracer uptake. RESULTS The co-registration between the autoradiographs and their corresponding histopathology was successful in all specimens. The use of this novel methodology allowed direct comparison of autoradiography and histopathology and enabled the visualisation of uncharted heterogeneity in [18F]FDG uptake in both benign and malignant tissue. CONCLUSION We here describe a novel methodology enabling the direct co-registration of [18F]FDG autoradiography with the gold standard of histopathology in human malignant tissue. The future use of the current methodology could further increase our understanding of the distribution of radionuclides in surgically excised malignancies and hence, improve the integration of pathology and molecular imaging in a multiscale perspective. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT05068687.
Collapse
Affiliation(s)
- Jens M Debacker
- Department of Head and Skin, Ghent University, Ghent, Belgium.
- Department of Head and Neck Surgery, Ghent University Hospital, Ghent, Belgium.
- Department of Nuclear Medicine, UZ Brussel, Brussels, Belgium.
- In vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium.
- Cancer Research Institute Ghent, Ghent, Belgium.
| | - Luna Maris
- Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
- XEOS Medical, Ghent, Belgium
| | - Fleur Cordier
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - David Creytens
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Philippe Deron
- Department of Head and Skin, Ghent University, Ghent, Belgium
- Department of Head and Neck Surgery, Ghent University Hospital, Ghent, Belgium
| | - Benedicte Descamps
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
- INFINITY Lab, Ghent University, Ghent, Belgium
| | - Yves D'Asseler
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Medical Imaging, Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| | - Kathia De Man
- Department of Medical Imaging, Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| | - Vincent Keereman
- Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
- XEOS Medical, Ghent, Belgium
| | - Sasha Libbrecht
- Department of Pathology, Antwerp University Hospital, Edegem, Belgium
| | - Vanessa Schelfhout
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Medical Imaging, Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| | - Koen Van de Vijver
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Christian Vanhove
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
- INFINITY Lab, Ghent University, Ghent, Belgium
| | - Wouter Huvenne
- Department of Head and Skin, Ghent University, Ghent, Belgium
- Department of Head and Neck Surgery, Ghent University Hospital, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
6
|
Piacentino F, Fontana F, Zorzetto G, Saccomanno A, Casagrande S, Franzi F, Imperatori A, Lanza C, Carriero S, Coppola A, Ierardi AM, Carrafiello G, Venturini M. Could Maximum SUV be Used as Imaging Guidance in Large Lung Lesions Biopsies? Double Sampling Under PET-CT/XperGuide Fusion Imaging in Inhomogeneous Lung Uptaking Lesions to Show That it can Make a Difference. Technol Cancer Res Treat 2023; 22:15330338221144508. [PMID: 37116886 PMCID: PMC10155026 DOI: 10.1177/15330338221144508] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023] Open
Abstract
Introduction: The purpose of this study is to evaluate the diagnostic value of positron emission computed tomography-cone beam computed tomography (PET/CT-CBCT) fusion guided percutaneous biopsy, targeted to the maximum standardized uptake value (SUVmax) and minimum standardized uptake value (SUVmin) of large lung lesions. Materials and Methods: Inside a larger cohort of PET/CT-CBCT guided percutaneous lung biopsies, 10 patients with large pulmonary lesions (diameter > 30 mm) were selected retrospectively. These patients have been subjected to double biopsy sampling respectively in the SUVmax area and in the SUVmin area of the lesion. Technical success has been calculated. For each sample, the percentage of neoplastic, inflammatory, and fibrotic cells was reported. Furthermore, the possibility of performing immunohistochemical or molecular biology investigations to specifically define the biomolecular tumor profile was analyzed. Results: Nine lesions were found to be malignant, one benign (inflammation). Technical success was 100% (10/10) in the SUVmax samples and 70% (7/10) in the SUVmin samples (P-value: .21). In the first group, higher percentages of neoplastic cells were found at pathologic evaluation, while in the second group areas of inflammation and fibrosis were more represented. The biomolecular profile was obtained in 100% of cases (9/9) of the first group, while in the second group only in 33.3% of cases (2/6), with a statistically significant difference between the 2 groups (P-value: .011). Conclusion: A correlation between the standardized uptake value value and the technical success of the biopsy sample has been identified. PET/CT-CBCT guidance allows to target the biopsy in the areas of the tumor which are richer in neoplastic cells, thus obtaining more useful information for the planning of patient-tailored cancer treatments.
Collapse
Affiliation(s)
- Filippo Piacentino
- Department of Diagnostic and Interventional Radiology, Circolo Hospital and Macchi Foundation, Insubria University, Varese, Italy
| | - Federico Fontana
- Department of Diagnostic and Interventional Radiology, Circolo Hospital and Macchi Foundation, Insubria University, Varese, Italy
| | - Giada Zorzetto
- Postgraduate School of Radiodiagnostics, Insubria University, Varese, Italy
| | - Angiola Saccomanno
- Postgraduate School of Radiodiagnostics, Insubria University, Varese, Italy
| | - Sabrina Casagrande
- Nuclear Medicine Unit, Circolo Hospital and Macchi Foundation, Varese, Italy
| | - Francesca Franzi
- Division of Pathological Anatomy, Circolo Hospital and Macchi Foundation, Insubria University, Varese, Italy
| | - Andrea Imperatori
- Division of Thoracic Surgery, Circolo Hospital and Macchi Foundation, Insubria University, Varese, Italy
| | - Carolina Lanza
- Postgraduate School of Radiodiagnostics, Università degli Studi di Milano, Milan, Italy
| | - Serena Carriero
- Postgraduate School of Radiodiagnostics, Università degli Studi di Milano, Milan, Italy
| | - Andrea Coppola
- Department of Diagnostic and Interventional Radiology, Circolo Hospital and Macchi Foundation, Insubria University, Varese, Italy
| | - Anna Maria Ierardi
- Interventional Radiology Unit, Department of Radiology, Foundation IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy
| | - Gianpaolo Carrafiello
- Interventional Radiology Unit, Department of Radiology, Foundation IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy
| | - Massimo Venturini
- Department of Diagnostic and Interventional Radiology, Circolo Hospital and Macchi Foundation, Insubria University, Varese, Italy
| |
Collapse
|
7
|
O'Donoghue J, Zanzonico P, Humm J, Kesner A. Dosimetry in Radiopharmaceutical Therapy. J Nucl Med 2022; 63:1467-1474. [PMID: 36192334 PMCID: PMC12079709 DOI: 10.2967/jnumed.121.262305] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 07/14/2022] [Indexed: 11/27/2022] Open
Abstract
The application of radiopharmaceutical therapy for the treatment of certain diseases is well established, and the field is expanding. New therapeutic radiopharmaceuticals have been developed in recent years, and more are in the research pipeline. Concurrently, there is growing interest in the use of internal dosimetry as a means of personalizing, and potentially optimizing, such therapy for patients. Internal dosimetry is multifaceted, and the current state of the art is discussed in this continuing education article. Topics include the context of dosimetry, internal dosimetry methods, the advantages and disadvantages of incorporating dosimetry calculations in radiopharmaceutical therapy, a description of the workflow for implementing patient-specific dosimetry, and future prospects in the field.
Collapse
Affiliation(s)
- Joe O'Donoghue
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pat Zanzonico
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - John Humm
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Adam Kesner
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
8
|
Hunt S, Zandifar A, Alavi A. Molecular imaging in management of colorectal metastases by the interventional oncologist. Int J Hyperthermia 2022; 39:675-681. [DOI: 10.1080/02656736.2021.1998657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Affiliation(s)
- Stephen Hunt
- Penn Image-Guided Interventions Lab, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Abass Alavi
- University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
9
|
Machine learning analysis for the noninvasive prediction of lymphovascular invasion in gastric cancer using PET/CT and enhanced CT-based radiomics and clinical variables. Abdom Radiol (NY) 2022; 47:1209-1222. [PMID: 35089370 DOI: 10.1007/s00261-021-03315-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 01/08/2023]
Abstract
PURPOSE Lymphovascular invasion (LVI) is associated with metastasis and poor survival in patients with gastric cancer, yet the noninvasive diagnosis of LVI is difficult. This study aims to develop predictive models using different machine learning (ML) classifiers based on both enhanced CT and PET/CT images and clinical variables for preoperatively predicting lymphovascular invasion (LVI) status of gastric cancer. METHODS A total of 101 patients with gastric cancer who underwent surgery were retrospectively recruited, and the LVI status was confirmed by pathological analysis. Patients were randomly divided into a training dataset (n = 76) and a validation dataset (n = 25). By 3D manual segmentation, radiomics features were extracted from the PET and venous phase CT images. Image models, clinical models, and combined models were constructed by selected enhanced CT-based and PET-based radiomics features, clinical factors, and a combination of both, respectively. Three ML classifiers including adaptive boosting (AdaBoost), linear discriminant analysis (LDA), and logistic regression (LR) were used for model development. The performance of these predictive models was evaluated with respect to discrimination, calibration, and clinical usefulness. RESULTS Ten radiomics features and eight clinical factors were selected for the development of predictive models. In the validation dataset, the area under curve (AUC) values of clinical models using AdaBoost, LDA, and LR classifiers were 0.742, 0.706, and 0.690, respectively. The image models using AdaBoost, LDA, and LR classifiers achieved an AUC of 0.849, 0.778, and 0.810, respectively. The combined models showed improved performance than the image models and the clinical models, with the AUC values of AdaBoost, LDA, and LR classifier yielding 0.944, 0.929, and 0.921, respectively. The combined models also showed good calibration and clinical usefulness for LVI prediction. CONCLUSION ML-based models integrating PET/CT and enhanced CT radiomics features and clinical factors have good discrimination capability, which could serve as a noninvasive, preoperative tool for the prediction of LVI and assist surgical treatment decisions in patients with gastric cancer.
Collapse
|
10
|
Anan N, Zainon R, Tamal M. A review on advances in 18F-FDG PET/CT radiomics standardisation and application in lung disease management. Insights Imaging 2022; 13:22. [PMID: 35124733 PMCID: PMC8817778 DOI: 10.1186/s13244-021-01153-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
Radiomics analysis quantifies the interpolation of multiple and invisible molecular features present in diagnostic and therapeutic images. Implementation of 18-fluorine-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) radiomics captures various disorders in non-invasive and high-throughput manner. 18F-FDG PET/CT accurately identifies the metabolic and anatomical changes during cancer progression. Therefore, the application of 18F-FDG PET/CT in the field of oncology is well established. Clinical application of 18F-FDG PET/CT radiomics in lung infection and inflammation is also an emerging field. Combination of bioinformatics approaches or textual analysis allows radiomics to extract additional information to predict cell biology at the micro-level. However, radiomics texture analysis is affected by several factors associated with image acquisition and processing. At present, researchers are working on mitigating these interrupters and developing standardised workflow for texture biomarker establishment. This review article focuses on the application of 18F-FDG PET/CT in detecting lung diseases specifically on cancer, infection and inflammation. An overview of different approaches and challenges encountered on standardisation of 18F-FDG PET/CT technique has also been highlighted. The review article provides insights about radiomics standardisation and application of 18F-FDG PET/CT in lung disease management.
Collapse
|
11
|
Sarrut D, Bała M, Bardiès M, Bert J, Chauvin M, Chatzipapas K, Dupont M, Etxebeste A, M Fanchon L, Jan S, Kayal G, S Kirov A, Kowalski P, Krzemien W, Labour J, Lenz M, Loudos G, Mehadji B, Ménard L, Morel C, Papadimitroulas P, Rafecas M, Salvadori J, Seiter D, Stockhoff M, Testa E, Trigila C, Pietrzyk U, Vandenberghe S, Verdier MA, Visvikis D, Ziemons K, Zvolský M, Roncali E. Advanced Monte Carlo simulations of emission tomography imaging systems with GATE. Phys Med Biol 2021; 66:10.1088/1361-6560/abf276. [PMID: 33770774 PMCID: PMC10549966 DOI: 10.1088/1361-6560/abf276] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/26/2021] [Indexed: 12/13/2022]
Abstract
Built on top of the Geant4 toolkit, GATE is collaboratively developed for more than 15 years to design Monte Carlo simulations of nuclear-based imaging systems. It is, in particular, used by researchers and industrials to design, optimize, understand and create innovative emission tomography systems. In this paper, we reviewed the recent developments that have been proposed to simulate modern detectors and provide a comprehensive report on imaging systems that have been simulated and evaluated in GATE. Additionally, some methodological developments that are not specific for imaging but that can improve detector modeling and provide computation time gains, such as Variance Reduction Techniques and Artificial Intelligence integration, are described and discussed.
Collapse
Affiliation(s)
- David Sarrut
- Université de Lyon, CREATIS, CNRS UMR5220, Inserm U1294, INSA-Lyon, Université Lyon 1, Lyon, France
| | | | - Manuel Bardiès
- Cancer Research Institute of Montpellier, U1194 INSERM/ICM/Montpellier University, 208 Av des Apothicaires, F-34298 Montpellier cedex 5, France
| | - Julien Bert
- LaTIM, INSERM UMR 1101, IBRBS, Faculty of Medicine, Univ Brest, 22 avenue Camille Desmoulins, F-29238, Brest, France
| | - Maxime Chauvin
- CRCT, UMR 1037, INSERM, Université Toulouse III Paul Sabatier, Toulouse, France
| | | | | | - Ane Etxebeste
- Université de Lyon, CREATIS, CNRS UMR5220, Inserm U1294, INSA-Lyon, Université Lyon 1, Lyon, France
| | - Louise M Fanchon
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, United States of America
| | - Sébastien Jan
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, F-91401, Orsay, France
| | - Gunjan Kayal
- CRCT, UMR 1037, INSERM, Université Toulouse III Paul Sabatier, Toulouse, France
- SCK CEN, Belgian Nuclear Research Centre, Boeretang 200, Mol 2400, Belgium
| | - Assen S Kirov
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, United States of America
| | - Paweł Kowalski
- High Energy Physics Division, National Centre for Nuclear Research, Otwock-Świerk, Poland
| | - Wojciech Krzemien
- High Energy Physics Division, National Centre for Nuclear Research, Otwock-Świerk, Poland
| | - Joey Labour
- Université de Lyon, CREATIS, CNRS UMR5220, Inserm U1294, INSA-Lyon, Université Lyon 1, Lyon, France
| | - Mirjam Lenz
- FH Aachen University of Applied Sciences, Forschungszentrum Jülich, Jülich, Germany
- Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - George Loudos
- Bioemission Technology Solutions (BIOEMTECH), Alexandras Av. 116, Athens, Greece
| | | | - Laurent Ménard
- Université Paris-Saclay, CNRS/IN2P3, IJCLab, F-91405 Orsay, France
- Université de Paris, IJCLab, F-91405 Orsay France
| | | | | | - Magdalena Rafecas
- Institute of Medical Engineering, University of Lübeck, Lübeck, Germany
| | - Julien Salvadori
- Department of Nuclear Medicine and Nancyclotep molecular imaging platform, CHRU-Nancy, Université de Lorraine, F-54000, Nancy, France
| | - Daniel Seiter
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, 53705, United States of America
| | - Mariele Stockhoff
- Medical Image and Signal Processing (MEDISIP), Ghent University, Ghent, Belgium
| | - Etienne Testa
- Univ. Lyon, Univ. Claude Bernard Lyon 1, CNRS/IN2P3, IP2I Lyon, F-69622, Villeurbanne, France
| | - Carlotta Trigila
- Department of Biomedical Engineering, University of California, Davis, CA 95616 United States of America
| | - Uwe Pietrzyk
- Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | | | - Marc-Antoine Verdier
- Université Paris-Saclay, CNRS/IN2P3, IJCLab, F-91405 Orsay, France
- Université de Paris, IJCLab, F-91405 Orsay France
| | - Dimitris Visvikis
- LaTIM, INSERM UMR 1101, IBRBS, Faculty of Medicine, Univ Brest, 22 avenue Camille Desmoulins, F-29238, Brest, France
| | - Karl Ziemons
- FH Aachen University of Applied Sciences, Forschungszentrum Jülich, Jülich, Germany
| | - Milan Zvolský
- Institute of Medical Engineering, University of Lübeck, Lübeck, Germany
| | - Emilie Roncali
- Department of Biomedical Engineering, University of California, Davis, CA 95616 United States of America
| |
Collapse
|
12
|
Intraperitoneal Glucose Transport to Micrometastasis: A Multimodal In Vivo Imaging Investigation in a Mouse Lymphoma Model. Int J Mol Sci 2021; 22:ijms22094431. [PMID: 33922728 PMCID: PMC8123046 DOI: 10.3390/ijms22094431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022] Open
Abstract
Bc-DLFL.1 is a novel spontaneous, high-grade transplantable mouse B-cell lymphoma model for selective serosal propagation. These cells attach to the omentum and mesentery and show dissemination in mesenteric lymph nodes. We aimed to investigate its early stage spread at one day post-intraperitoneal inoculation of lymphoma cells (n = 18 mice), and its advanced stage at seven days post-inoculation with in vivo [18F]FDG-PET and [18F]PET/MRI, and ex vivo by autoradiography and Cherenkov luminescence imaging (CLI). Of the early stage group, nine animals received intraperitoneal injections, and nine received intravenous [18F]FDG injections. The advanced stage group (n = 3) received intravenous FDG injections. In the early stage, using autoradiography we observed a marked accumulation in the mesentery after intraperitoneal FDG injection. Using other imaging methods and autoradiography, following the intravenous injection of FDG no accumulations were detected. At the advanced stage, tracer accumulation was clearly detected in mesenteric lymph nodes and in the peritoneum after intravenous administration using PET. We confirmed the results with immunohistochemistry. Our results in this model highlight the importance of local FDG administration during diagnostic imaging to precisely assess early peritoneal manifestations of other malignancies (colon, stomach, ovary). These findings also support the importance of applying topical therapies, in addition to systemic treatments in peritoneal cancer spread.
Collapse
|
13
|
Khan S, Kim S, Yang YP, Pratx G. High-resolution radioluminescence microscopy of FDG uptake in an engineered 3D tumor-stoma model. Eur J Nucl Med Mol Imaging 2021; 48:3400-3407. [PMID: 33880604 DOI: 10.1007/s00259-021-05364-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 04/07/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE The increased glucose metabolism of cancer cells is the basis for 18F-fluorodeoxyglucose positron emission tomography (FDG-PET). However, due to its coarse image resolution, PET is unable to resolve the metabolic role of cancer-associated stroma, which often influences the metabolic reprogramming of a tumor. This study investigates the use of radioluminescence microscopy for imaging FDG uptake in engineered 3D tumor models with high resolution. METHOD Multicellular tumor spheroids (A549 lung adenocarcinoma) were co-cultured with GFP-expressing human umbilical vein endothelial cells (HUVECs) within an artificial extracellular matrix to mimic a tumor and its surrounding stroma. The tumor model was constructed as a 200-μm-thin 3D layer over a transparent CdWO4 scintillator plate to allow high-resolution imaging of the cultured cells. After incubation with FDG, the radioluminescence signal was collected by a highly sensitive widefield microscope. Fluorescence microscopy was performed using the same instrument to localize endothelial and tumor cells. RESULTS Simultaneous and co-localized brightfield, fluorescence, and radioluminescence imaging provided high-resolution information on the distribution of FDG in the engineered tissue. The microvascular stromal compartment as a whole took up a large fraction of the FDG, comparable to the uptake of the tumor spheroids. In vitro gamma counting confirmed that A549 and HUVEC cells were both highly glycolytic with rapid FDG uptake kinetics. Despite the relative thickness of the tissue constructs, an average spatial resolution of 64 ± 4 μm was achieved for imaging FDG. CONCLUSION Our study demonstrates the feasibility of imaging the distribution of FDG uptake in engineered in vitro tumor models. With its high spatial resolution, the method can separately resolve tumor and stromal components. The approach could be extended to more advanced engineered cancer models but also to surgical tissue slices and tumor biopsies.
Collapse
Affiliation(s)
- Syamantak Khan
- Department of Radiation Oncology, Stanford University, Stanford, CA, 94305, USA
| | - Sungwoo Kim
- Department of Orthopedic Surgery, Stanford University, Stanford, CA, 94305, USA
| | - Yunzhi Peter Yang
- Department of Orthopedic Surgery, Stanford University, Stanford, CA, 94305, USA
| | - Guillem Pratx
- Department of Radiation Oncology, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
14
|
Bogoni M, Cerci JJ, Cornelis FH, Nanni C, Tabacchi E, SchÖder H, Shyn PB, Sofocleous CT, Solomon SB, Kirov AS. Practice and prospects for PET/CT guided interventions. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2021; 65:20-31. [PMID: 33494585 PMCID: PMC10446123 DOI: 10.23736/s1824-4785.21.03291-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
During the past 10 years, performing real-time molecular imaging with positron emission tomography (PET) in combination with computed tomography (CT) during interventional procedures has undergone rapid development. Keeping in mind the interest of the nuclear medicine readers, an update is provided of the current workflows using real-time PET/CT in percutaneous biopsies and tumor ablations. The clinical utility of PET/CT guided biopsies in cancer patients with lung, liver, lymphoma, and bone tumors are reviewed. Several technological developments, including the introduction of new PET tracers and robotic arms as well as opportunities provided through acquiring radioactive biopsy specimens are briefly reviewed.
Collapse
Affiliation(s)
| | | | | | - Cristina Nanni
- Unit of Nuclear Medicine, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Elena Tabacchi
- Unit of Nuclear Medicine, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Heiko SchÖder
- Unit of Nuclear Medicine, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paul B Shyn
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Constantinos T Sofocleous
- Unit of Interventional Radiology, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stephen B Solomon
- Unit of Interventional Radiology, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Assen S Kirov
- Unit of Molecular Imaging and Therapy Physics, Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA -
| |
Collapse
|
15
|
Tselikas L, Champiat S, Sheth RA, Yevich S, Ammari S, Deschamps F, Farhane S, Roux C, Susini S, Mouraud S, Delpla A, Raoult T, Robert C, Massard C, Barlesi F, Soria JC, Marabelle A, de Baere T. Interventional Radiology for Local Immunotherapy in Oncology. Clin Cancer Res 2021; 27:2698-2705. [PMID: 33419781 DOI: 10.1158/1078-0432.ccr-19-4073] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/24/2020] [Accepted: 01/04/2021] [Indexed: 11/16/2022]
Abstract
Human intratumoral immunotherapy (HIT-IT) is under rapid development, with promising preliminary results and high expectations for current phase III trials. While outcomes remain paramount for patients and the referring oncologists, the technical aspects of drug injection are critical to the interventional radiologist to ensure optimal and reproducible outcomes. The technical considerations for HIT-IT affect the safety, efficacy, and further development of this treatment option. Image-guided access to the tumor allows the therapeutic index of a treatment to be enhanced by increasing the intratumoral drug concentration while minimizing its systemic exposure and associated on-target off-tumor adverse events. Direct access to the tumor also enables the acquisition of cancer tissue for sequential sampling to better understand the pharmacodynamics of the injected immunotherapy and its efficacy through correlation of immune responses, pathologic responses, and imaging tumor response. The aim of this article is to share the technical insights of HIT-IT, with particular consideration for patient selection, lesion assessment, image guidance, and technical injection options. In addition, the organization of a standard patient workflow is discussed, so as to optimize HIT-IT outcome and the patient experience.
Collapse
Affiliation(s)
- Lambros Tselikas
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Gustave Roussy, Villejuif, France. .,Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Gustave Roussy, Villejuif, France
| | - Stephane Champiat
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Rahul A Sheth
- Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steve Yevich
- Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samy Ammari
- Radiology Department, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Frederic Deschamps
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Gustave Roussy, Villejuif, France
| | - Siham Farhane
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Charles Roux
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Gustave Roussy, Villejuif, France
| | - Sandrine Susini
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Gustave Roussy, Villejuif, France
| | - Severine Mouraud
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Gustave Roussy, Villejuif, France
| | - Alexandre Delpla
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Gustave Roussy, Villejuif, France
| | - Thibault Raoult
- Service de Promotion des Etudes Cliniques (SPEC), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Caroline Robert
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France.,Gustave Roussy Cancer Campus, Université Paris Saclay, Saint-Aubin, France
| | - Christophe Massard
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France.,Gustave Roussy Cancer Campus, Université Paris Saclay, Saint-Aubin, France
| | - Fabrice Barlesi
- Gustave Roussy Cancer Campus, Université Paris Saclay, Saint-Aubin, France
| | - Jean-Charles Soria
- Gustave Roussy Cancer Campus, Université Paris Saclay, Saint-Aubin, France
| | - Aurélien Marabelle
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Gustave Roussy, Villejuif, France.,Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Thierry de Baere
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Gustave Roussy, Villejuif, France.,Gustave Roussy Cancer Campus, Université Paris Saclay, Saint-Aubin, France
| |
Collapse
|
16
|
Fontana F, Piacentino F, Ierardi AM, Carrafiello G, Coppola A, Muollo A, Beneventi A, Floridi C, Imperatori AS, Carcano G, Venturini M. Comparison Between CBCT and Fusion PET/CT-CBCT Guidance for Lung Biopsies. Cardiovasc Intervent Radiol 2020; 44:73-79. [PMID: 32895781 DOI: 10.1007/s00270-020-02613-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 08/02/2020] [Indexed: 12/23/2022]
Abstract
PURPOSE To establish the feasibility of performing percutaneous biopsy of lung lesions guided by fusion PET/CT-CBCT and to evaluate whether the metabolic information provided by a prior PET/CT scan add incremental benefits for diagnosis. METHODS We retrospectively reviewed data from 180 patients who underwent CBCT-guided lung biopsy (group 1-90 cases) or PET/CT-CBCT fusion-guided lung biopsy (group 2-90 cases). Technical and clinical success was calculated. We also evaluated the agreement between biopsy and definitive histology and the possibility to carrying out immunehistochemical and molecular biology analyses. RESULTS Technical success was achieved in 84/90 (93.3%) cases for group 1 and 89/90 (98.9%) for group 2 cases (p 0.054). Clinical success was achieved in 80/94 (95.2%) cases for group 1 and 88/89 (98.9%) cases for group 2. Sensitivity, specificity, positive and negative predictive values and accuracy rate were, respectively, 94.5%, 100.0%, 100.0%, 73.3% and 95.2% for group 1 and 98.6%, 100.0%, 100.0%, 94.4% and 98.9% for group 2 (p 0.167). Agreement between biopsy and definitive histology was reached in 85.7% for group 1 and in 96.2% for group 2 (p 0.211). Immunohistochemical and molecular biology investigations were possible in 66.7% for group 1 and in 77.0% for group 2 (p 0.297). No major complication occurred. CONCLUSIONS PET/CT-CBCT-guided lung biopsy is a feasible technique. In our retrospective case series, we found a higher clinical success rate, but no statistical difference was found.
Collapse
Affiliation(s)
- Federico Fontana
- Diagnostic and Interventional Radiology Department, Ospedale di Circolo e Fondazione Macchi, ASST dei Sette Laghi, Via Guicciardini, 9, 21100, Varese, Italy
- Università degli Studi dell'Insubria, Varese, Italy
| | - Filippo Piacentino
- Diagnostic and Interventional Radiology Department, Ospedale di Circolo e Fondazione Macchi, ASST dei Sette Laghi, Via Guicciardini, 9, 21100, Varese, Italy
| | - Anna Maria Ierardi
- Diagnostic and Interventional Radiology Department, ASST Santi Paolo e Carlo, San Paolo Hospital, Milan, Italy
- Università degli Studi di Milano, Milan, Italy
| | - Gianpaolo Carrafiello
- Diagnostic and Interventional Radiology Department, ASST Santi Paolo e Carlo, San Paolo Hospital, Milan, Italy
- Università degli Studi di Milano, Milan, Italy
| | - Andrea Coppola
- Diagnostic and Interventional Radiology Department, Ospedale di Circolo e Fondazione Macchi, ASST dei Sette Laghi, Via Guicciardini, 9, 21100, Varese, Italy.
| | - Alessandra Muollo
- Radiology Department, Ospedale di Legnano, ASST Ovest Milanese, Legnano, MI, Italy
| | - Alessandro Beneventi
- Diagnostic and Interventional Radiology Department, Ospedale Sant'Anna, ASST Lariana, Como, Italy
| | | | - Andrea Selenito Imperatori
- Università degli Studi dell'Insubria, Varese, Italy
- Thoracic Surgery Department, Ospedale di Circolo e Fondazione Macchi, ASST dei Sette Laghi, Varese, Italy
| | - Giulio Carcano
- Università degli Studi dell'Insubria, Varese, Italy
- Surgery Department, Ospedale di Circolo e Fondazione Macchi, ASST dei Sette Laghi, Varese, Italy
| | - Massimo Venturini
- Diagnostic and Interventional Radiology Department, Ospedale di Circolo e Fondazione Macchi, ASST dei Sette Laghi, Via Guicciardini, 9, 21100, Varese, Italy
- Università degli Studi dell'Insubria, Varese, Italy
| |
Collapse
|
17
|
Kishore SA, Drabkin MJ, Sofocleous CT. Fluorodeoxyglucose-PET for Ablation Treatment Planning, Intraprocedural Monitoring, and Response. PET Clin 2020; 14:427-436. [PMID: 31472740 DOI: 10.1016/j.cpet.2019.06.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PET has become an essential tool for staging and response assessment in oncologic imaging. Over the past decade it has also evolved into a tool for image-guided interventions, specifically in the rapidly growing field of interventional oncology. PET-guided biopsies have greater sensitivity and diagnostic yield for fluorodeoxyglucose-avid lesions. Real-time PET imaging can also provide valuable image guidance during therapeutic minimally invasive procedures such as ablation of PET-avid tumors. The increasing use of PET in the assessment of therapeutic response results in earlier identification of disease that is amenable to image-guided therapies.
Collapse
Affiliation(s)
- Sirish A Kishore
- Interventional Radiology Service, Memorial Sloan Kettering Cancer, 1275 York, IR Suite H118, New York City, NY 10065, USA
| | - Michael J Drabkin
- Interventional Radiology Service, Memorial Sloan Kettering Cancer, New York City, NY, USA
| | - Constantinos T Sofocleous
- Interventional Radiology Service, Memorial Sloan Kettering Cancer, 1275 York, IR Suite H118, New York City, NY 10065, USA.
| |
Collapse
|
18
|
Percutaneous Lung Tumor Biopsy Under CBCT Guidance with PET-CT Fusion Imaging: Preliminary Experience. Cardiovasc Intervent Radiol 2019; 42:1644-1648. [DOI: 10.1007/s00270-019-02270-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 06/13/2019] [Indexed: 12/11/2022]
|
19
|
Kirov AS, Fanchon LM, Seiter D, Czmielewski C, Russell J, Dogan S, Carlin S, Pinker-Domenig K, Yorke E, Schmidtlein CR, Boyko V, Fujisawa S, Manova-Todorova K, Zanzonico P, Dauer L, Deasy JO, Humm JL, Solomon S. Technical Note: Scintillation well counters and particle counting digital autoradiography devices can be used to detect activities associated with genomic profiling adequacy of biopsy specimens obtained after a low activity 18 F-FDG injection. Med Phys 2018; 45:2179-2185. [PMID: 29480927 DOI: 10.1002/mp.12836] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 02/14/2018] [Accepted: 02/15/2018] [Indexed: 11/08/2022] Open
Abstract
PURPOSE Genomic profiling of biopsied tissue is the basis for precision cancer therapy. However, biopsied materials may not contain sufficient amounts of tumor deoxyribonucleonic acid needed for the analysis. We propose a method to determine the adequacy of specimens for performing genomic profiling by quantifying their metabolic activity. METHODS We estimated the average density of tumor cells in biopsy specimens needed to successfully perform genomic analysis following the Memorial Sloan Kettering Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT) protocol from the minimum amount of deoxyribonucleonic acid needed and the volume of tissue typically used for analysis. The average 18 F-FDG uptake per cell was assessed by incubating HT-29 adenocarcinoma tumor cells in 18 F-FDG containing solution and then measuring their activity with a scintillation well counter. Consequently, we evaluated the response of two devices around the minimum expected activities which would indicate genomic profiling adequacy of biopsy specimens obtained under 18 F-FDG PET/CT guidance. Surrogate samples obtained using 18G core needle biopsies of gels containing either 18 F-FDG-loaded cells in the expected concentrations or the corresponding activity were measured using autoradiography and a scintillation well counter. Autoradiography was performed using a CCD-based device with real-time image display as well as with digital autoradiography imaging plates following a 30-min off-line protocol for specimen activity determination against previously established calibration. RESULTS Cell incubation experiments and estimates obtained from quantitative autoradiography of biopsy specimens (QABS) indicate that specimens acquired under 18 F-FDG PET/CT guidance that contained the minimum amount of cells needed for genomic profiling would have an average activity concentration in the range of about 3 to about 9 kBq/mL. When exposed to specimens with similar activity concentration, both a CCD-based autoradiography device and a scintillation well counter produced signals with sufficient signal-to-background ratio for specimen genomic adequacy identification in less than 10 min, which is short enough to allow procedure guidance. CONCLUSION Scintillation well counter measurements and CCD-based autoradiography have adequate sensitivity to detect the tumor burden needed for genomic profiling during 18 F-FDG PET/CT-guided 18G core needle biopsies of liver adenocarcinoma metastases.
Collapse
Affiliation(s)
- Assen S Kirov
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Louise M Fanchon
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | | | - Christian Czmielewski
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - James Russell
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Snjezana Dogan
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Sean Carlin
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Katja Pinker-Domenig
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Ellen Yorke
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - C Ross Schmidtlein
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Vitaly Boyko
- Molecular Cytology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Sho Fujisawa
- Molecular Cytology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Katia Manova-Todorova
- Molecular Cytology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Pat Zanzonico
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Lawrence Dauer
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Joseph O Deasy
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - John L Humm
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Stephen Solomon
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| |
Collapse
|
20
|
Lu M, Zhan X. The crucial role of multiomic approach in cancer research and clinically relevant outcomes. EPMA J 2018; 9:77-102. [PMID: 29515689 PMCID: PMC5833337 DOI: 10.1007/s13167-018-0128-8] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 01/29/2018] [Indexed: 02/06/2023]
Abstract
Cancer with heavily economic and social burden is the hot point in the field of medical research. Some remarkable achievements have been made; however, the exact mechanisms of tumor initiation and development remain unclear. Cancer is a complex, whole-body disease that involves multiple abnormalities in the levels of DNA, RNA, protein, metabolite and medical imaging. Biological omics including genomics, transcriptomics, proteomics, metabolomics and radiomics aims to systematically understand carcinogenesis in different biological levels, which is driving the shift of cancer research paradigm from single parameter model to multi-parameter systematical model. The rapid development of various omics technologies is driving one to conveniently get multi-omics data, which accelerates predictive, preventive and personalized medicine (PPPM) practice allowing prediction of response with substantially increased accuracy, stratification of particular patients and eventual personalization of medicine. This review article describes the methodology, advances, and clinically relevant outcomes of different "omics" technologies in cancer research, and especially emphasizes the importance and scientific merit of integrating multi-omics in cancer research and clinically relevant outcomes.
Collapse
Affiliation(s)
- Miaolong Lu
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
| | - Xianquan Zhan
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
- The State Key Laboratory of Medical Genetics, Central South University, 88 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
| |
Collapse
|
21
|
Fanchon LM, Apte A, Schmidtlein CR, Yorke E, Hu YC, Dogan S, Hatt M, Visvikis D, Humm JL, Solomon SB, Kirov AS. Evaluation of the tumor registration error in biopsy procedures performed under real-time PET/CT guidance. Med Phys 2017; 44:5089-5095. [PMID: 28494089 DOI: 10.1002/mp.12334] [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: 11/01/2016] [Revised: 04/21/2017] [Accepted: 04/21/2017] [Indexed: 12/15/2022] Open
Abstract
PURPOSE The purpose of this study is to quantify tumor displacement during real-time PET/CT guided biopsy and to investigate correlations between tumor displacement and false-negative results. METHODS 19 patients who underwent real-time 18 F-FDG PET-guided biopsy and were found positive for malignancy were included in this study under IRB approval. PET/CT images were acquired for all patients within minutes prior to biopsy to visualize the FDG-avid region and plan the needle insertion. The biopsy needle was inserted and a post-insertion CT scan was acquired. The two CT scans acquired before and after needle insertion were registered using a deformable image registration (DIR) algorithm. The DIR deformation vector field (DVF) was used to calculate the mean displacement between the pre-insertion and post-insertion CT scans for a region around the tip of the biopsy needle. For 12 patients one biopsy core from each was tracked during histopathological testing to investigate correlations of the mean displacement between the two CT scans and false-negative or true-positive biopsy results. For 11 patients, two PET scans were acquired; one at the beginning of the procedure, pre-needle insertion, and an additional one with the needle in place. The pre-insertion PET scan was corrected for intraprocedural motion by applying the DVF. The corrected PET was compared with the post-needle insertion PET to validate the correction method. RESULTS The mean displacement of tissue around the needle between the pre-biopsy CT and the postneedle insertion CT was 5.1 mm (min = 1.1 mm, max = 10.9 mm and SD = 3.0 mm). For mean displacements larger than 7.2 mm, the biopsy cores gave false-negative results. Correcting pre-biopsy PET using the DVF improved the PET/CT registration in 8 of 11 cases. CONCLUSIONS The DVF obtained from DIR of the CT scans can be used for evaluation and correction of the error in needle placement with respect to the FDG-avid area. Misregistration between the pre-biopsy PET and the CT acquired with the needle in place was shown to correlate with false negative biopsy results.
Collapse
Affiliation(s)
- Louise M Fanchon
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Adytia Apte
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - C Ross Schmidtlein
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Ellen Yorke
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Yu-Chi Hu
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Snjezana Dogan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Mathieu Hatt
- INSERM, UMR 1101, LaTIM, IBRBS, UBO, Brest, France
| | | | - John L Humm
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Stephen B Solomon
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Assen S Kirov
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| |
Collapse
|
22
|
Hatt M, Lee JA, Schmidtlein CR, Naqa IE, Caldwell C, De Bernardi E, Lu W, Das S, Geets X, Gregoire V, Jeraj R, MacManus MP, Mawlawi OR, Nestle U, Pugachev AB, Schöder H, Shepherd T, Spezi E, Visvikis D, Zaidi H, Kirov AS. Classification and evaluation strategies of auto-segmentation approaches for PET: Report of AAPM task group No. 211. Med Phys 2017; 44:e1-e42. [PMID: 28120467 DOI: 10.1002/mp.12124] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 12/09/2016] [Accepted: 01/04/2017] [Indexed: 12/14/2022] Open
Abstract
PURPOSE The purpose of this educational report is to provide an overview of the present state-of-the-art PET auto-segmentation (PET-AS) algorithms and their respective validation, with an emphasis on providing the user with help in understanding the challenges and pitfalls associated with selecting and implementing a PET-AS algorithm for a particular application. APPROACH A brief description of the different types of PET-AS algorithms is provided using a classification based on method complexity and type. The advantages and the limitations of the current PET-AS algorithms are highlighted based on current publications and existing comparison studies. A review of the available image datasets and contour evaluation metrics in terms of their applicability for establishing a standardized evaluation of PET-AS algorithms is provided. The performance requirements for the algorithms and their dependence on the application, the radiotracer used and the evaluation criteria are described and discussed. Finally, a procedure for algorithm acceptance and implementation, as well as the complementary role of manual and auto-segmentation are addressed. FINDINGS A large number of PET-AS algorithms have been developed within the last 20 years. Many of the proposed algorithms are based on either fixed or adaptively selected thresholds. More recently, numerous papers have proposed the use of more advanced image analysis paradigms to perform semi-automated delineation of the PET images. However, the level of algorithm validation is variable and for most published algorithms is either insufficient or inconsistent which prevents recommending a single algorithm. This is compounded by the fact that realistic image configurations with low signal-to-noise ratios (SNR) and heterogeneous tracer distributions have rarely been used. Large variations in the evaluation methods used in the literature point to the need for a standardized evaluation protocol. CONCLUSIONS Available comparison studies suggest that PET-AS algorithms relying on advanced image analysis paradigms provide generally more accurate segmentation than approaches based on PET activity thresholds, particularly for realistic configurations. However, this may not be the case for simple shape lesions in situations with a narrower range of parameters, where simpler methods may also perform well. Recent algorithms which employ some type of consensus or automatic selection between several PET-AS methods have potential to overcome the limitations of the individual methods when appropriately trained. In either case, accuracy evaluation is required for each different PET scanner and scanning and image reconstruction protocol. For the simpler, less robust approaches, adaptation to scanning conditions, tumor type, and tumor location by optimization of parameters is necessary. The results from the method evaluation stage can be used to estimate the contouring uncertainty. All PET-AS contours should be critically verified by a physician. A standard test, i.e., a benchmark dedicated to evaluating both existing and future PET-AS algorithms needs to be designed, to aid clinicians in evaluating and selecting PET-AS algorithms and to establish performance limits for their acceptance for clinical use. The initial steps toward designing and building such a standard are undertaken by the task group members.
Collapse
Affiliation(s)
- Mathieu Hatt
- INSERM, UMR 1101, LaTIM, University of Brest, IBSAM, Brest, France
| | - John A Lee
- Université catholique de Louvain (IREC/MIRO) & FNRS, Brussels, 1200, Belgium
| | | | | | - Curtis Caldwell
- Sunnybrook Health Sciences Center, Toronto, ON, M4N 3M5, Canada
| | | | - Wei Lu
- Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Shiva Das
- University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Xavier Geets
- Université catholique de Louvain (IREC/MIRO) & FNRS, Brussels, 1200, Belgium
| | - Vincent Gregoire
- Université catholique de Louvain (IREC/MIRO) & FNRS, Brussels, 1200, Belgium
| | - Robert Jeraj
- University of Wisconsin, Madison, WI, 53705, USA
| | | | | | - Ursula Nestle
- Universitätsklinikum Freiburg, Freiburg, 79106, Germany
| | - Andrei B Pugachev
- University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Heiko Schöder
- Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | | | - Emiliano Spezi
- School of Engineering, Cardiff University, Cardiff, Wales, United Kingdom
| | | | - Habib Zaidi
- Geneva University Hospital, Geneva, CH-1211, Switzerland
| | - Assen S Kirov
- Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| |
Collapse
|
23
|
Abstract
OBJECTIVE The purpose of this review is to summarize the applications of PET molecular imaging-directed biopsy of a variety of organs in the management of various diseases with a focus on cancers. CONCLUSION PET can yield metabolic information at the cellular and molecular levels, and PET-directed biopsy is playing an increasing role in the diagnosis and staging of diseases.
Collapse
|
24
|
Agarwal KK, Behera A, Kumar R, Bal C. 18F-Fluorodeoxyglucose-Positron Emission Tomography/Computed Tomography in Tuberculosis: Spectrum of Manifestations. Indian J Nucl Med 2017; 32:316-321. [PMID: 29142348 PMCID: PMC5672752 DOI: 10.4103/ijnm.ijnm_29_17] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The objective of this article is to provide an illustrative tutorial highlighting the utility of 18F-fluorodeoxyglucose-positron emission tomography/computed tomography (18F-FDG-PET/CT) imaging to detect spectrum of manifestations in patients with tuberculosis (TB). FDG-PET/CT is a powerful tool for early diagnosis, measuring the extent of disease (staging), and consequently for evaluation of response to therapy in patients with TB.
Collapse
Affiliation(s)
- Krishan Kant Agarwal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Abhishek Behera
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Rakesh Kumar
- Department of Nuclear Medicine, Division of Diagnostic Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Chandrasekhar Bal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
25
|
Wright CL, Maly JJ, Zhang J, Knopp MV. Advancing Precision Nuclear Medicine and Molecular Imaging for Lymphoma. PET Clin 2016; 12:63-82. [PMID: 27863567 DOI: 10.1016/j.cpet.2016.08.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PET with fluorodeoxyglucose F 18 (18F FDG-PET) is a meaningful biomarker for the detection, targeted biopsy, and treatment of lymphoma. This article reviews the evolution of 18F FDG-PET as a putative biomarker for lymphoma and addresses the current capabilities, challenges, and opportunities to enable precision medicine practices for lymphoma. Precision nuclear medicine is driven by new imaging technologies and methodologies to more accurately detect malignant disease. Although quantitative assessment of response is limited, such technologies will enable a more precise metabolic mapping with much higher definition image detail and thus may make it a robust and valid quantitative response assessment methodology.
Collapse
Affiliation(s)
- Chadwick L Wright
- Wright Center of Innovation in Biomedical Imaging, Division of Imaging Science, Department of Radiology, The Ohio State University Wexner Medical Center, 395 West 12th Avenue, Room 430, Columbus, OH 43210, USA
| | - Joseph J Maly
- Division of Hematology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Starling Loving Hall 406C, 320 West 10th Avenue, Columbus, OH 43210, USA
| | - Jun Zhang
- Wright Center of Innovation in Biomedical Imaging, Division of Imaging Science, Department of Radiology, The Ohio State University Wexner Medical Center, 395 West 12th Avenue, Room 430, Columbus, OH 43210, USA
| | - Michael V Knopp
- Wright Center of Innovation in Biomedical Imaging, Division of Imaging Science, Department of Radiology, The Ohio State University Wexner Medical Center, 395 West 12th Avenue, Room 430, Columbus, OH 43210, USA.
| |
Collapse
|
26
|
Abstract
Radiomics is an emerging field in quantitative imaging that uses advanced imaging features to objectively and quantitatively describe tumour phenotypes. Radiomic features have recently drawn considerable interest due to its potential predictive power for treatment outcomes and cancer genetics, which may have important applications in personalized medicine. In this technical review, we describe applications and challenges of the radiomic field. We will review radiomic application areas and technical issues, as well as proper practices for the designs of radiomic studies.
Collapse
Affiliation(s)
- Stephen S F Yip
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
27
|
Maybody M, Grewal RK, Healey JH, Antonescu CR, Fanchon L, Hwang S, Carrasquillo JA, Kirov A, Farooki A. Ga-68 DOTATOC PET/CT-Guided Biopsy and Cryoablation with Autoradiography of Biopsy Specimen for Treatment of Tumor-Induced Osteomalacia. Cardiovasc Intervent Radiol 2016; 39:1352-7. [PMID: 27150801 DOI: 10.1007/s00270-016-1350-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/08/2016] [Indexed: 10/21/2022]
Abstract
Tumor-induced osteomalacia (TIO) is a rare paraneoplastic syndrome caused by small benign tumors of mesenchymal origin also known as phosphaturic mesenchymal tumors mixed connective tissue variant. Excellent prognosis is expected with eradication of the culprit tumor. These small tumors are notoriously difficult to localize with conventional imaging studies; this often leads to an extensive work up and prolonged morbidity. We report a patient with clinical diagnosis of TIO whose culprit tumor was localized with Ga-68 DOTATOC PET/CT and MRI. Biopsy and cryoablation were performed under Ga-68 DOTATOC PET/CT guidance. Autoradiography of the biopsy specimen was performed and showed in situ correlation between Ga-68 DOTATOC uptake and histopathology with millimeter resolution.
Collapse
Affiliation(s)
- Majid Maybody
- Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, M276C, New York, NY, 10065, USA.
| | - Ravinder K Grewal
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John H Healey
- Orthopedic Surgical Oncology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cristina R Antonescu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Louise Fanchon
- Department of Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sinchun Hwang
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jorge A Carrasquillo
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Assen Kirov
- Department of Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Azeez Farooki
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
28
|
Abstract
PET can be used to guide percutaneous needle biopsy to the most metabolic lesion, improving diagnostic yield. PET biopsy guidance can be performed using visual or software coregistration, electromagnetic needle tracking, cone-beam computed tomography (CT), and intraprocedural PET/CT guidance. PET/CT-guided biopsies allow the sampling of lesions that may not be clearly visible on anatomic imaging, or of lesions that are morphologically normal. PET can identify suspicious locations within complex tumors that are most likely to contain important diagnostic and prognostic information.
Collapse
Affiliation(s)
- Ghassan El-Haddad
- Division of Interventional Radiology, Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612-9416, USA.
| |
Collapse
|
29
|
Abstract
Although molecular imaging has had a dramatic impact on diagnostic imaging, it has only recently begun to be integrated into interventional procedures. Its significant impact is attributed to its ability to provide noninvasive, physiologic information that supplements conventional morphologic imaging. The four major interventional opportunities for molecular imaging are, first, to provide guidance to localize a target; second, to provide tissue analysis to confirm that the target has been reached; third, to provide in-room, posttherapy assessment; and fourth, to deliver targeted therapeutics. With improved understanding and application of(18)F-FDG, as well as the addition of new molecular probes beyond(18)F-FDG, the future holds significant promise for the expansion of molecular imaging into the realm of interventional procedures.
Collapse
Affiliation(s)
- Stephen B Solomon
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Francois Cornelis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York; and Department of Radiology, Pellegrin Hospital, Bordeaux, France
| |
Collapse
|