1
|
Mordi A, Karunakaran V, Marium Mim U, Marple E, Rajaram N. Design and Validation of a Multimodal Diffuse Reflectance and Spatially Offset Raman Spectroscopy System for In Vivo Applications. JOURNAL OF BIOPHOTONICS 2025; 18:e202400333. [PMID: 39721981 PMCID: PMC11884968 DOI: 10.1002/jbio.202400333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024]
Abstract
We report on the development of a multimodal spectroscopy system, combining diffuse reflectance spectroscopy (DRS) and spatially offset Raman spectroscopy (SORS). A fiber optic probe was designed with spatially offset source-detector fibers to collect subsurface measurements for each modality, as well as ball lens-coupled fibers for superficial measurements. The system acquires DRS, zero-offset Raman spectroscopy (RS) and SORS with good signal-to-noise ratio. Measurements on chicken breast tissue demonstrate that both DRS and RS can acquire spectra from similar depths within tissue. Measurements acquired from the skin of a human volunteer demonstrate distinct Raman peaks at 937 and 1755 cm-1 that were unique to the zero-offset ball lens configuration and 718 and 1089 cm-1 for the spatially offset setting. We also identified Raman peaks corresponding to melanin that were prominent in the superficial measurements obtained with the ball lens-coupled fibers but not in the spatially offset fibers.
Collapse
Affiliation(s)
- April Mordi
- Department of Biomedical EngineeringUniversity of ArkansasFayettevilleArkansasUSA
| | - Varsha Karunakaran
- Department of Biomedical EngineeringUniversity of ArkansasFayettevilleArkansasUSA
| | - Umme Marium Mim
- Department of Biomedical EngineeringUniversity of ArkansasFayettevilleArkansasUSA
| | | | - Narasimhan Rajaram
- Department of Biomedical EngineeringUniversity of ArkansasFayettevilleArkansasUSA
| |
Collapse
|
2
|
Shen X, Zhou H, Zhou X, Liu Z, Meng X, Zhang L, Song Y, Guo R, Wang F, Li K, Li W, Yang Z, Liu Z, Li N. 68Ga-grazytracer PET for noninvasive assessment of response to immunotherapy in solid tumors and lymphomas: a phase 1/2 clinical trial. Nat Commun 2024; 15:8791. [PMID: 39389969 PMCID: PMC11467221 DOI: 10.1038/s41467-024-53197-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024] Open
Abstract
To tackle the clinical challenge of noninvasively assessing immunotherapy efficacy in patients, here we used positron emission tomography (PET) with 68Ga-grazytracer, which targets granzyme B, a crucial effector molecule secreted by activated CD8+ T cells. In this phase 1/2 clinical trial (NCT05000372) involving a diverse cohort of 24 patients with solid tumors and lymphomas who received immunotherapies, including immune checkpoint inhibitors (either alone or with chemotherapies) and chimeric antigen receptor-T cell therapy, we examined the in vivo behaviors of 68Ga-grazytracer. Primary endpoints were safety, biodistribution, granzyme B specificity, and the predictive utility of 68Ga-grazytracer, while secondary endpoint was the relationship between 68Ga-grazytracer uptake and tumor immune phenotype. 68Ga-grazytracer exhibited a safe profile and specifically targeted granzyme B in patients. 68Ga-grazytracer PET showed superior predictive value for short-term prognosis and progression-free survival than those of conventional assessment criteria, including RECIST 1.1 and PERCIST. Moreover, the uptake of 68Ga-grazytracer in tumors was significantly higher in those with a "non-desert" immune phenotype than those with an immune "desert" phenotype, thereby meeting the primary and secondary endpoints of this trial. Collectively, we successfully visualized CD8+ T cell effector function in humans using 68Ga-grazytracer PET, offering insights for enhancing immunotherapy assessment, patient stratification and treatment planning.
Collapse
Affiliation(s)
- Xiuling Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Haoyi Zhou
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Xin Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Zongchao Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Xiangxi Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Linyu Zhang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Yufei Song
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Rui Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Fei Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Kui Li
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Wenqing Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 100142, Beijing, China.
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, 100142, Beijing, China.
| | - Zhaofei Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 100142, Beijing, China.
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, 100142, Beijing, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China.
- Department of Nuclear Medicine, Peking University Third Hospital, 100191, Beijing, China.
| | - Nan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 100142, Beijing, China.
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, 100142, Beijing, China.
| |
Collapse
|
3
|
Sunassee ED, Jardim-Perassi BV, Madonna MC, Ordway B, Ramanujam N. Metabolic Imaging as a Tool to Characterize Chemoresistance and Guide Therapy in Triple-Negative Breast Cancer (TNBC). Mol Cancer Res 2023; 21:995-1009. [PMID: 37343066 PMCID: PMC10592445 DOI: 10.1158/1541-7786.mcr-22-1004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 04/07/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023]
Abstract
After an initial response to chemotherapy, tumor relapse is frequent. This event is reflective of both the spatiotemporal heterogeneities of the tumor microenvironment as well as the evolutionary propensity of cancer cell populations to adapt to variable conditions. Because the cause of this adaptation could be genetic or epigenetic, studying phenotypic properties such as tumor metabolism is useful as it reflects molecular, cellular, and tissue-level dynamics. In triple-negative breast cancer (TNBC), the characteristic metabolic phenotype is a highly fermentative state. However, during treatment, the spatial and temporal dynamics of the metabolic landscape are highly unstable, with surviving populations taking on a variety of metabolic states. Thus, longitudinally imaging tumor metabolism provides a promising approach to inform therapeutic strategies, and to monitor treatment responses to understand and mitigate recurrence. Here we summarize some examples of the metabolic plasticity reported in TNBC following chemotherapy and review the current metabolic imaging techniques available in monitoring chemotherapy responses clinically and preclinically. The ensemble of imaging technologies we describe has distinct attributes that make them uniquely suited for a particular length scale, biological model, and/or features that can be captured. We focus on TNBC to highlight the potential of each of these technological advances in understanding evolution-based therapeutic resistance.
Collapse
Affiliation(s)
- Enakshi D. Sunassee
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | | | - Megan C. Madonna
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Bryce Ordway
- Department of Cancer Physiology, Moffitt Cancer Center, Tampa, FL 33612, USA
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Nirmala Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27708, USA
| |
Collapse
|
4
|
Ma X, Mao M, He J, Liang C, Xie HY. Nanoprobe-based molecular imaging for tumor stratification. Chem Soc Rev 2023; 52:6447-6496. [PMID: 37615588 DOI: 10.1039/d3cs00063j] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
The responses of patients to tumor therapies vary due to tumor heterogeneity. Tumor stratification has been attracting increasing attention for accurately distinguishing between responders to treatment and non-responders. Nanoprobes with unique physical and chemical properties have great potential for patient stratification. This review begins by describing the features and design principles of nanoprobes that can visualize specific cell types and biomarkers and release inflammatory factors during or before tumor treatment. Then, we focus on the recent advancements in using nanoprobes to stratify various therapeutic modalities, including chemotherapy, radiotherapy (RT), photothermal therapy (PTT), photodynamic therapy (PDT), chemodynamic therapy (CDT), ferroptosis, and immunotherapy. The main challenges and perspectives of nanoprobes in cancer stratification are also discussed to facilitate probe development and clinical applications.
Collapse
Affiliation(s)
- Xianbin Ma
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Mingchuan Mao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Jiaqi He
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Chao Liang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Hai-Yan Xie
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Chemical Biology Center, Peking University, Beijing, 100191, P. R. China.
| |
Collapse
|
5
|
Cutshaw G, Hassan N, Uthaman S, Wen X, Singh B, Sarkar A, Bardhan R. Monitoring Metabolic Changes in Response to Chemotherapies in Cancer with Raman Spectroscopy and Metabolomics. Anal Chem 2023; 95:13172-13184. [PMID: 37605298 PMCID: PMC10845238 DOI: 10.1021/acs.analchem.3c02073] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
Resistance to clinical therapies remains a major barrier in cancer management. There is a critical need for rapid and highly sensitive diagnostic tools that enable early prediction of treatment response to allow accurate clinical decisions. Here, Raman spectroscopy was employed to monitor changes in key metabolites as early predictors of response in KRAS-mutant colorectal cancer (CRC) cells, HCT116, treated with chemotherapies. We show at the single cell level that HCT116 is resistant to cetuximab (CTX), the first-line treatment in CRC, but this resistance can be overcome with pre-sensitization of cells with oxaliplatin (OX). In combination treatment of CTX + OX, sequential delivery of OX followed by CTX rather than simultaneous administration of drugs was observed to be critical for effective therapy. Our results demonstrated that metabolic changes are well aligned to cellular mechanical changes where Young's modulus decreased after effective treatment, indicating that both changes in mechanical properties and metabolism in cells are likely responsible for cancer proliferation. Raman findings were verified with mass spectrometry (MS) metabolomics, and both platforms showed changes in lipids, nucleic acids, and amino acids as predictors of resistance/response. Finally, key metabolic pathways enriched were identified when cells are resistant to CTX but downregulated with effective treatment. This study highlights that drug-induced metabolic changes both at the single cell level (Raman) and ensemble level (MS) have the potential to identify mechanisms of response to clinical cancer therapies.
Collapse
Affiliation(s)
- Gabriel Cutshaw
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA
- Nanovaccine Institute, Iowa State University, Ames, IA 50012, USA
| | - Nora Hassan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA
- Nanovaccine Institute, Iowa State University, Ames, IA 50012, USA
| | - Saji Uthaman
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA
- Nanovaccine Institute, Iowa State University, Ames, IA 50012, USA
| | - Xiaona Wen
- Nanovaccine Institute, Iowa State University, Ames, IA 50012, USA
| | - Bhuminder Singh
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Anwesha Sarkar
- Department of Electrical Engineering, Iowa State University, Ames, IA 50012, USA
| | - Rizia Bardhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA
- Nanovaccine Institute, Iowa State University, Ames, IA 50012, USA
| |
Collapse
|
6
|
Khalil D, Lotfalla A, Girard A, Ha R, Dercle L, Seban RD. Advances in PET/CT Imaging for Breast Cancer Patients and Beyond. J Clin Med 2023; 12:jcm12020651. [PMID: 36675588 PMCID: PMC9861174 DOI: 10.3390/jcm12020651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/19/2023] Open
Abstract
Breast cancer is the most common cancer in women around the world and the fifth leading cause of cancer-related death [...].
Collapse
Affiliation(s)
- David Khalil
- Campbell University School of Osteopathic Medicine, Lillington, NC 27546, USA
| | - Andrew Lotfalla
- Touro College of Osteopathic Medicine, Middletown, NY 10940, USA
| | - Antoine Girard
- Department of Nuclear Medicine, CHU Amiens-Picardie, 80000 Amiens, France
| | - Richard Ha
- Department of Radiology, NewYork-Presbyterian, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Laurent Dercle
- Department of Radiology, NewYork-Presbyterian, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Romain-David Seban
- Department of Nuclear Medicine, Institut Curie, 92210 Saint-Cloud, France
- Correspondence:
| |
Collapse
|
7
|
Pac J, Koo DJ, Cho H, Jung D, Choi MH, Choi Y, Kim B, Park JU, Kim SY, Lee Y. Three-dimensional imaging and analysis of pathological tissue samples with de novo generation of citrate-based fluorophores. SCIENCE ADVANCES 2022; 8:eadd9419. [PMID: 36383671 PMCID: PMC9668299 DOI: 10.1126/sciadv.add9419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/29/2022] [Indexed: 06/16/2023]
Abstract
Two-dimensional (2D) histopathology based on the observation of thin tissue slides is the current paradigm in diagnosis and prognosis. However, labeling strategies in conventional histopathology are limited in compatibility with 3D imaging combined with tissue clearing techniques. Here, we present a rapid and efficient volumetric imaging technique of pathological tissues called 3D tissue imaging through de novo formation of fluorophores, or 3DNFC, which is the integration of citrate-based fluorogenic reaction DNFC and tissue clearing techniques. 3DNFC markedly increases the fluorescence intensity of tissues by generating fluorophores on nonfluorescent amino-terminal cysteine and visualizes the 3D structure of the tissues to provide their anatomical morphology and volumetric information. Furthermore, the application of 3DNFC to pathological tissue achieves the 3D reconstruction for the unbiased analysis of diverse features of the disorders in their natural context. We suggest that 3DNFC is a promising volumetric imaging method for the prognosis and diagnosis of pathological tissues.
Collapse
Affiliation(s)
- Jinyoung Pac
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
| | - Dong-Jun Koo
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, South Korea
| | - Hyeongjun Cho
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, South Korea
| | - Dongwook Jung
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
| | - Min-ha Choi
- Department of Plastic and Reconstructive Surgery, Seoul National University Boramae Hospital, Seoul National University College of Medicine, 5 Gil 20, Boramae Road, Dongjak-Gu, Seoul 07061, South Korea
| | - Yunjung Choi
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
| | - Bohyun Kim
- Department of Pathology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, South Korea
| | - Ji-Ung Park
- Department of Plastic and Reconstructive Surgery, Seoul National University Boramae Hospital, Seoul National University College of Medicine, 5 Gil 20, Boramae Road, Dongjak-Gu, Seoul 07061, South Korea
| | - Sung-Yon Kim
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, South Korea
| | - Yan Lee
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
| |
Collapse
|
8
|
Challenges and opportunities in the development of metal-based anticancer theranostic agents. Biosci Rep 2022; 42:231168. [PMID: 35420649 PMCID: PMC9109461 DOI: 10.1042/bsr20212160] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/07/2022] [Accepted: 04/13/2022] [Indexed: 12/02/2022] Open
Abstract
Around 10 million fatalities were recorded worldwide in 2020 due to cancer and statistical projections estimate the number to increase by 60% in 2040. With such a substantial rise in the global cancer burden, the disease will continue to impose a huge socio-economic burden on society. Currently, the most widely used clinical treatment modality is cytotoxic chemotherapy using platinum drugs which is used to treat variety of cancers. Despite its clinical success, critical challenges like resistance, off-target side effects and cancer variability often reduce its overall therapeutic efficiency. These challenges require faster diagnosis, simultaneous therapy and a more personalized approach toward cancer management. To this end, small-molecule ‘theranostic’ agents have presented a viable solution combining diagnosis and therapy into a single platform. In this review, we present a summary of recent efforts in the design and optimization of metal-based small-molecule ‘theranostic’ anticancer agents. Importantly, we highlight the advantages of a theranostic candidate over the purely therapeutic or diagnostic agent in terms of evaluation of its biological properties.
Collapse
|
9
|
PET imaging in breast cancer. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00124-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
10
|
Kwon HW, Lee JH, Pahk K, Park KH, Kim S. Clustering subtypes of breast cancer by combining immunohistochemistry profiles and metabolism characteristics measured using FDG PET/CT. Cancer Imaging 2021; 21:55. [PMID: 34579791 PMCID: PMC8477513 DOI: 10.1186/s40644-021-00424-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 09/07/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The aim of this study was to investigate the effect of combining immunohistochemical profiles and metabolic information to characterize breast cancer subtypes. METHODS This retrospective study included 289 breast tumors from 284 patients who underwent preoperative 18 F-fluorodeoxyglucose (FDG) positron emission tomography/ computed tomography (PET/CT). Molecular subtypes of breast cancer were classified as Hormonal, HER2, Dual (a combination of both Hormonal and HER2 features), and triple-negative (TN). Histopathologic findings and immunohistochemical results for Ki-67, EGFR, CK 5/6, and p53 were also analyzed. The maximum standardized uptake value (SUV) measured from FDG PET/CT was used to evaluate tumoral glucose metabolism. RESULTS Overall, 182, 24, 47, and 36 tumors were classified as Hormonal, HER2, Dual, and TN subtypes, respectively. Molecular profiles of tumor aggressiveness and the tumor SUV revealed a gradual increase from the Hormonal to the TN type. The tumor SUV was significantly correlated with tumor size, expression levels of p53, Ki-67, and EGFR, and nuclear grade (all p < 0.001). In contrast, the tumor SUV was negatively correlated with the expression of estrogen receptors (r = - 0.234, p < 0.001) and progesterone receptors (r = - 0.220, p < 0.001). Multiple linear regression analysis revealed that histopathologic markers explained tumor glucose metabolism (adjusted R-squared value 0.238, p < 0.001). Tumor metabolism can thus help define breast cancer subtypes with aggressive/adverse prognostic features. CONCLUSIONS Metabolic activity measured using FDG PET/CT was significantly correlated with the molecular alteration profiles of breast cancer assessed using immunohistochemical analysis. Combining molecular markers and metabolic information may aid in the recognition and understanding of tumor aggressiveness in breast cancer and be helpful as a prognostic marker.
Collapse
Affiliation(s)
- Hyun Woo Kwon
- Department of Nuclear Medicine, Korea University College of Medicine, Seoul, Korea
| | - Jeong Hyeon Lee
- Department of Pathology, Korea University College of Medicine, Seoul, Korea
| | - Kisoo Pahk
- Department of Nuclear Medicine, Korea University College of Medicine, Seoul, Korea
| | - Kyong Hwa Park
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Sungeun Kim
- Department of Nuclear Medicine, Korea University College of Medicine, Seoul, Korea.
- Department of Nuclear Medicine, Korea University Anam Hospital, Korea University College of Medicine, 73 Goryeodae-ro, Seongbuk-gu, 02841, Seoul, Korea.
| |
Collapse
|
11
|
Capozza M, Anemone A, Dhakan C, Della Peruta M, Bracesco M, Zullino S, Villano D, Terreno E, Longo DL, Aime S. GlucoCEST MRI for the Evaluation Response to Chemotherapeutic and Metabolic Treatments in a Murine Triple-Negative Breast Cancer: A Comparison with[ 18F]F-FDG-PET. Mol Imaging Biol 2021; 24:126-134. [PMID: 34383241 DOI: 10.1007/s11307-021-01637-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/30/2021] [Accepted: 07/28/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) patients have usually poor outcome after chemotherapy and early prediction of therapeutic response would be helpful. [18F]F-FDG-PET/CT acquisitions are often carried out to monitor variation in metabolic activity associated with response to the therapy, despite moderate accuracy and radiation exposure limit its application. The glucoCEST technique relies on the use of unlabelled D-glucose to assess glucose uptake with conventional MRI scanners and is currently under active investigations at clinical level. This work aims at validating the potential of MRI-glucoCEST in monitoring the therapeutic responses in a TNBC tumor murine model. PROCEDURES Breast tumor (4T1)-bearing mice were treated with doxorubicin or dichloroacetate for 1 week. PET/CT with [18F]F-FDG and MRI-glucoCEST were performed at baseline and after 3 cycles of treatment. Metabolic changes measured with [18F]F-FDG-PET and glucoCEST were compared and evaluated with changes in tumor volumes. RESULTS Doxorubicin-treated mice showed a significant decrease in tumor growth when compared to the control group. GlucoCEST imaging provided metabolic response after three cycles of treatment. Conversely, no variations were detected in [18F]F-FDG uptake. Dichloroacetate-treated mice did not show any decrease either in tumor volume or in tumor metabolic activity as assessed by both glucoCEST and [18F]F-FDG-PET. CONCLUSIONS Metabolic changes during doxorubicin treatment can be predicted by glucoCEST imaging that appears more sensitive than [18F]F-FDG-PET in reporting on therapeutic response. These findings support the view that glucoCEST may be a sensitive technique for monitoring metabolic response, but future studies are needed to explore the accuracy of this approach in other tumor types and treatments.
Collapse
Affiliation(s)
- Martina Capozza
- Center for Preclinical Imaging, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, Turin, 10126, Italy
| | - Annasofia Anemone
- Molecular Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, Turin, 10126, Italy
| | - Chetan Dhakan
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, Turin, 10126, Italy
| | - Melania Della Peruta
- Center for Preclinical Imaging, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, Turin, 10126, Italy
| | - Martina Bracesco
- Center for Preclinical Imaging, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, Turin, 10126, Italy
| | - Sara Zullino
- Molecular Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, Turin, 10126, Italy
| | - Daisy Villano
- Molecular Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, Turin, 10126, Italy
| | - Enzo Terreno
- Center for Preclinical Imaging, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, Turin, 10126, Italy.,Molecular Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, Turin, 10126, Italy.,Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, Turin, 10126, Italy
| | - Dario Livio Longo
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, Turin, 10126, Italy
| | - Silvio Aime
- Molecular Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, Turin, 10126, Italy.,Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, Turin, 10126, Italy
| |
Collapse
|
12
|
Nakata J, Isohashi K, Oka Y, Nakajima H, Morimoto S, Fujiki F, Oji Y, Tsuboi A, Kumanogoh A, Hashimoto N, Hatazawa J, Sugiyama H. Imaging Assessment of Tumor Response in the Era of Immunotherapy. Diagnostics (Basel) 2021; 11:diagnostics11061041. [PMID: 34198874 PMCID: PMC8226723 DOI: 10.3390/diagnostics11061041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/29/2021] [Accepted: 06/03/2021] [Indexed: 02/07/2023] Open
Abstract
Assessment of tumor response during treatment is one of the most important purposes of imaging. Before the appearance of immunotherapy, response evaluation criteria in solid tumors (RECIST) and positron emission tomography response criteria in solid tumors (PERCIST) were, respectively, the established morphologic and metabolic response criteria, and cessation of treatment was recommended when progressive disease was detected according to these criteria. However, various types of immunotherapy have been developed over the past 20 years, which show novel false positive findings on images, as well as distinct response patterns from conventional therapies. Antitumor immune response itself causes 18F-fluorodeoxyglucose (FDG) uptake in tumor sites, known as "flare phenomenon", so that positron emission tomography using FDG can no longer accurately identify remaining tumors. Furthermore, tumors often initially increase, followed by stability or decrease resulting from immunotherapy, which is called "pseudoprogression", so that progressive disease cannot be confirmed by computed tomography or magnetic resonance imaging at a single time point. As a result, neither RECIST nor PERCIST can accurately predict the response to immunotherapy, and therefore several new response criteria fixed for immunotherapy have been proposed. However, these criteria are still controversial, and also require months for response confirmation. The establishment of optimal response criteria and the development of new imaging technologies other than FDG are therefore urgently needed. In this review, we summarize the false positive images and the revision of response criteria for each immunotherapy, in order to avoid discontinuation of a truly effective immunotherapy.
Collapse
Affiliation(s)
- Jun Nakata
- Department of Clinical Laboratory and Biomedical Sciences, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan;
- Correspondence: ; Tel.: +81-6-6879-3676; Fax: +81-6-6879-3677
| | - Kayako Isohashi
- Department of Kansai BNCT Medical Center, Osaka Medical and Pharmaceutical University, Takatsuki City 596-8686, Osaka, Japan;
| | - Yoshihiro Oka
- Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan;
- Department of Immunopathology, WP1 Immunology Frontier Research Center, Osaka University, Suita City 565-0871, Osaka, Japan;
| | - Hiroko Nakajima
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan; (H.N.); (F.F.); (H.S.)
| | - Soyoko Morimoto
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan; (S.M.); (A.T.)
| | - Fumihiro Fujiki
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan; (H.N.); (F.F.); (H.S.)
| | - Yusuke Oji
- Department of Clinical Laboratory and Biomedical Sciences, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan;
| | - Akihiro Tsuboi
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan; (S.M.); (A.T.)
| | - Atsushi Kumanogoh
- Department of Immunopathology, WP1 Immunology Frontier Research Center, Osaka University, Suita City 565-0871, Osaka, Japan;
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan
| | - Naoya Hashimoto
- Department of Neurosurgery, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto City 602-8566, Kyoto, Japan;
| | - Jun Hatazawa
- Department of Research Center for Nuclear Physics, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan;
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan; (H.N.); (F.F.); (H.S.)
| |
Collapse
|
13
|
Huang Y, Le J, Miao A, Zhi W, Wang F, Chen Y, Zhou S, Chang C. Prediction of treatment responses to neoadjuvant chemotherapy in breast cancer using contrast-enhanced ultrasound. Gland Surg 2021; 10:1280-1290. [PMID: 33968680 DOI: 10.21037/gs-20-836] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Elucidation the efficacy of neoadjuvant chemotherapy (NAC) in breast cancer is important for informing therapeutic decisions. This study aimed at evaluating the potential value of contrast-enhanced ultrasound (CEUS) parameters in predicting breast cancer responses to NAC. Methods We performed CEUS examinations before and after two cycles of NAC. Quantitative CEUS parameters [maximum intensity (IMAX), rise time (RT), time to peak (TTP), and mean transit time (mTT)], tumor diameter, and their changes were measured and compared to histopathological responses, according to the Miller-Payne Grading (MPG) system (score 1, 2, or 3: minor response; score 4 or 5: good response). Prediction models for good response were developed by multiple logistic regression analysis and internally validated through bootstrap analysis. The receiver operating characteristic (ROC) curve was used to evaluate the performance of prediction models. Results A total of 143 patients were enrolled in this study among whom 98 (68.5%) achieved a good response and while 45 (31.5%) exhibited a minor response. Several imaging variables including diameter, IMAX, changes in diameter (Δdiameter), IMAX (ΔIMAX) and TTP (ΔTTP) were found to be significantly associated with good therapeutic responses (P<0.05). The areas under the curve (AUC) increased from 0.748 to 0.841 in the multivariate model that combined CEUS parameters and molecular subtypes with a sensitivity and specificity of 0.786, 0.745, respectively. Tumor molecular subtype was the primary predictor of primary endpoint. Conclusions CEUS is a potential tool for predicting responses to NAC in locally advanced breast cancer patients. Compared to the other molecular subtypes, triple negative and HER2+/ER- subtypes are more likely to exhibit a good response to NAC.
Collapse
Affiliation(s)
- Yunxia Huang
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Le
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Aiyu Miao
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenxiang Zhi
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fen Wang
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yaling Chen
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shichong Zhou
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cai Chang
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
14
|
Tribukait B. Early prediction of pathologic response to neoadjuvant treatment of breast cancer: use of a cell-loss metric based on serum thymidine kinase 1 and tumour volume. BMC Cancer 2020; 20:440. [PMID: 32423477 PMCID: PMC7236455 DOI: 10.1186/s12885-020-06925-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 05/03/2020] [Indexed: 01/08/2023] Open
Abstract
Background After neoadjuvant chemotherapy of breast cancer pathologic complete response (pCR) indicates a favorable prognosis. Among non-selected patients, pCR is, however, achieved in only 10–30%. Early evaluation of tumour response to treatment would facilitate individualized therapy, with ineffective chemotherapy interrupted or changed. The methodology for this purpose is still limited. Tumour imaging and analysis of macromolecules, released from disrupted tumour cells, are principal alternatives. Objective To investigate whether a metric of cell-loss, defined as the ratio between serum concentration of thymidine kinase1 (sTK1, ng x ml− 1) and tumour volume, can be used for early prediction of pathologic response. Methods One hunred four women with localized breast cancer received neoadjuvant epirubicin/docetaxel in 6 cycles, supplemented with bevacizumab in cycles 3–6. The cell-loss metric was established at baseline (n = 104), 48 h after cycle 2 (n = 104) and prior to cycle 2 (n = 57). The performance of the metric was evaluated by association with pathologic tumour response at surgery 4 months later. Results Treatment caused a rise in sTK1, a reduction in tumour volume and a marked increase in the cell-loss metric. Patients were subdivided into quartiles according to the baseline cell-loss metric. For these groups, baseline means were 0.0016, 0.0042, 0.0062, 0.0178 units. After subtraction of baselines, means for the quartiles 48 h after treatment 2 were 0.002, 0.011, 0.030 and 0.357 units. pCR was achieved in 24/104, their distribution in the quartiles (11, 11, 23 and 46%) differed significantly (p = 0.01). In 80 patients with remaining tumour, tumour size was inversely related to the metric (p = 0.002). In 57 patients studied before treatment 2, positive and negative predictive values of the metric were 77.8 and 83.3%, compared to 40.5 and 88.7% 48 h after treatment 2. Conclusion A cell-loss metric, based on serum levels of TK1, released from disrupted tumour cells, and tumour volume, reveal tumour response early during neoadjuvant treatment. The metric reflect tumour properties that differ greatly between patients and determine the sensitivity to cytotoxic treatment. The findings point to the significance of cell loss for tumour growth rate. The metric should be considered in personalized oncology and in the evaluation of new therapeutic modalities. Trial registration PROMIX (Clinical Trials.govNCT000957125).
Collapse
Affiliation(s)
- Bernhard Tribukait
- Department of Oncology-Pathology, Karolinska Institute and University Hospital Solna, Stockholm, Sweden. .,Cancer Centrum Karolinska, CCK, Plan 00, Visionsgatan 56, Karolinska Universitetssjukhuset, Solna, 17164, Stockholm, Sweden.
| |
Collapse
|
15
|
Tribukait B. Early prediction of pathologic response to neoadjuvant treatment of breast cancer: use of a cell-loss metric based on serum thymidine kinase 1 and tumour volume. BMC Cancer 2020. [PMID: 32423477 DOI: 10.1186/s12885-020-06925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND After neoadjuvant chemotherapy of breast cancer pathologic complete response (pCR) indicates a favorable prognosis. Among non-selected patients, pCR is, however, achieved in only 10-30%. Early evaluation of tumour response to treatment would facilitate individualized therapy, with ineffective chemotherapy interrupted or changed. The methodology for this purpose is still limited. Tumour imaging and analysis of macromolecules, released from disrupted tumour cells, are principal alternatives. OBJECTIVE To investigate whether a metric of cell-loss, defined as the ratio between serum concentration of thymidine kinase1 (sTK1, ng x ml- 1) and tumour volume, can be used for early prediction of pathologic response. METHODS One hunred four women with localized breast cancer received neoadjuvant epirubicin/docetaxel in 6 cycles, supplemented with bevacizumab in cycles 3-6. The cell-loss metric was established at baseline (n = 104), 48 h after cycle 2 (n = 104) and prior to cycle 2 (n = 57). The performance of the metric was evaluated by association with pathologic tumour response at surgery 4 months later. RESULTS Treatment caused a rise in sTK1, a reduction in tumour volume and a marked increase in the cell-loss metric. Patients were subdivided into quartiles according to the baseline cell-loss metric. For these groups, baseline means were 0.0016, 0.0042, 0.0062, 0.0178 units. After subtraction of baselines, means for the quartiles 48 h after treatment 2 were 0.002, 0.011, 0.030 and 0.357 units. pCR was achieved in 24/104, their distribution in the quartiles (11, 11, 23 and 46%) differed significantly (p = 0.01). In 80 patients with remaining tumour, tumour size was inversely related to the metric (p = 0.002). In 57 patients studied before treatment 2, positive and negative predictive values of the metric were 77.8 and 83.3%, compared to 40.5 and 88.7% 48 h after treatment 2. CONCLUSION A cell-loss metric, based on serum levels of TK1, released from disrupted tumour cells, and tumour volume, reveal tumour response early during neoadjuvant treatment. The metric reflect tumour properties that differ greatly between patients and determine the sensitivity to cytotoxic treatment. The findings point to the significance of cell loss for tumour growth rate. The metric should be considered in personalized oncology and in the evaluation of new therapeutic modalities. TRIAL REGISTRATION PROMIX (Clinical Trials.govNCT000957125).
Collapse
Affiliation(s)
- Bernhard Tribukait
- Department of Oncology-Pathology, Karolinska Institute and University Hospital Solna, Stockholm, Sweden. .,Cancer Centrum Karolinska, CCK, Plan 00, Visionsgatan 56, Karolinska Universitetssjukhuset, Solna, 17164, Stockholm, Sweden.
| |
Collapse
|
16
|
Gao W, Guo N, Dong T. Diffusion-weighted imaging in monitoring the pathological response to neoadjuvant chemotherapy in patients with breast cancer: a meta-analysis. World J Surg Oncol 2018; 16:145. [PMID: 30021656 PMCID: PMC6052572 DOI: 10.1186/s12957-018-1438-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 06/26/2018] [Indexed: 01/22/2023] Open
Abstract
Background Diffusion-weighted imaging (DWI) is suggested as an non-invasive and non-radioactive imaging modality in the identification of pathological complete response (pCR) in breast cancer patients receiving neoadjuvant chemotherapy (NACT). A growing number of trials have been investigating in this aspect and some studies found a superior performance of DWI compared with conventional imaging techniques. However, the efficiency of DWI is still in dispute. This meta-analysis aims at evaluating the accuracy of DWI in the detection of pCR to NACT in patients with breast cancer. Methods Pooled sensitivity, specificity, and diagnostic odds ratio (DOR) were drawn to estimate the diagnostic effect of DWI to NACT. Summary receiver operating characteristic curve (SROC), the area under the SROC curve (AUC), and Youden index (*Q) were also calculated. The possible sources of heterogeneity among the included studies were explored using single-factor meta-regression analyses. Publication bias and quality assessment were assessed using Deek’s funnel plot and QUADAS-2 form respectively. Results Twenty studies incorporated 1490 participants were enrolled in our analysis. Pooled estimates revealed a sensitivity of 0.89 (95% CI, 0.86–0.91), a specificity of 0.72 (95% CI, 0.68–0.75), and a DOR of 27.00 (95% CI, 15.60–46.73). The AUC of SROC curve and *Q index were 0.9088 and 0.8408, respectively. The results of meta-regression analyses showed that pCR rate, time duration of study population, and study design were not the sources of heterogeneity. Conclusion A relatively high sensitivity and specificity of DWI in diagnosing pCP for patients with breast cancer underwent NACT treatment was found in our meta-analysis. This finding indicated that the use of DWI might provide an accurate and precise assessment of pCR to NACT.
Collapse
Affiliation(s)
- Wen Gao
- Department of Trauma Surgery, Tianjin Fourth Central Hospital, No.1 Zhongshan Road, Hebei District, Tianjin, 300010, China
| | - Ning Guo
- Department of Breast Surgery, Tianjin Fourth Central Hospital, No.1 Zhongshan Road, Hebei District, Tianjin, 300010, China
| | - Ting Dong
- Department of Cardiovascular Medicine, Guizhou Provincial People's Hospital, No. 83 Zhongshandong Road, Guiyang City, 550002, Guizhou, China.
| |
Collapse
|
17
|
Factors Impacting Pathologic Complete Response after Neoadjuvant Chemotherapy in Breast Cancer: A Single-Center Study. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2018. [DOI: 10.5812/ijcm.60098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
18
|
Didonato R, Shapiro N, Koenigsberg T, D'Alfonso T, Jaffer S, Fineberg S. Invasive mucinous carcinoma of the breast and response patterns after neoadjuvant chemotherapy (NAC). Histopathology 2018; 72:965-973. [DOI: 10.1111/his.13451] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 12/03/2017] [Indexed: 11/28/2022]
Affiliation(s)
- Rosemarie Didonato
- Department of Pathology; Montefiore Medical Center and the Albert Einstein College of Medicine; Bronx NY USA
| | - Nella Shapiro
- Montefiore Medical Center and The Eastchester Center for Cancer Care; Bronx NY USA
| | - Tova Koenigsberg
- Division of Breast Imaging; Montefiore Medical Center and the Albert Einstein College of Medicine; Bronx NY USA
| | - Timothy D'Alfonso
- Weill Cornell Medical Center and Cornell Medical College; New York NY USA
| | - Shabnam Jaffer
- Breast Pathology; Mount Sinai Medical Center; New York NY USA
| | - Susan Fineberg
- Department of Pathology; Montefiore Medical Center and the Albert Einstein College of Medicine; Bronx NY USA
| |
Collapse
|
19
|
Ueda S, Saeki T, Osaki A, Yamane T, Kuji I. Bevacizumab Induces Acute Hypoxia and Cancer Progression in Patients with Refractory Breast Cancer: Multimodal Functional Imaging and Multiplex Cytokine Analysis. Clin Cancer Res 2017; 23:5769-5778. [PMID: 28679773 DOI: 10.1158/1078-0432.ccr-17-0874] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/29/2017] [Accepted: 06/30/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Bevacizumab, an antibody against endothelial growth factor, is a key but controversial drug in the treatment of metastatic breast cancer. We, therefore, aimed to determine the intrinsic resistance to bevacizumab at the physiologic and molecular levels in advanced breast cancer using PET, dynamic contrast-enhanced MRI, diffuse optical spectroscopic imaging (DOSI), and multiplex cytokine assays.Experimental Design: In total, 28 patients diagnosed with advanced stage III/IV breast cancer receiving single-agent bevacizumab for 1 week followed by paclitaxel combined with bevacizumab underwent 18F-fluorodeoxyglucose (FDG)-PET, 18F-fluoromisonidazole (FMISO)-PET, and MRI at both baseline and two courses after treatment initiation. Hemodynamic measurement using DOSI and blood sample collection were performed at baseline and multiple times during the first week after the initiation of single-agent bevacizumab. We distinguished nonresponders from responders by serial FDG-PET based on their glycolytic changes to chemotherapy.Results: Nonresponders showed significantly higher hypoxic activity on FMISO-PET and less tumor shrinkage than responders. Hemodynamic parameters showed higher tumor blood volume and a remarkable decrease in the tissue oxygen level in nonresponders compared with responders after the infusion of single-agent bevacizumab. Multiplex cytokine assays revealed increased plasma levels of both proangiogenic and hypoxia-related inflammatory cytokines in nonresponders and decreased levels in responders.Conclusions: Nonresponders exhibited a higher degree of angiogenesis with more severe hypoxia than responders during bevacizumab treatment. These findings demonstrated that the addition of bevacizumab to paclitaxel treatment under hypoxic conditions could be ineffective and may result in acute hypoxia and increased cytokine secretion associated with cancer progression. Clin Cancer Res; 23(19); 5769-78. ©2017 AACR.
Collapse
Affiliation(s)
- Shigeto Ueda
- Department of Breast Oncology, Saitama Medical University International Medical Center, Yamane, Hidaka, Saitama, Japan.
| | - Toshiaki Saeki
- Department of Breast Oncology, Saitama Medical University International Medical Center, Yamane, Hidaka, Saitama, Japan
| | - Akihiko Osaki
- Department of Breast Oncology, Saitama Medical University International Medical Center, Yamane, Hidaka, Saitama, Japan
| | - Tomohiko Yamane
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, Yamane, Hidaka, Saitama, Japan
| | - Ichiei Kuji
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, Yamane, Hidaka, Saitama, Japan
| |
Collapse
|
20
|
Paganelli G, Matteucci F, Gilardi L. Nuclear Medicine in the Clinical Management (ROLL, SNB, and PET). Breast Cancer 2017. [DOI: 10.1007/978-3-319-48848-6_19] [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]
|
21
|
Comparison of WHO, RECIST 1.1, EORTC, and PERCIST criteria in the evaluation of treatment response in malignant solid tumors. Nucl Med Commun 2016; 37:9-15. [PMID: 26440568 DOI: 10.1097/mnm.0000000000000401] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
AIM To compare response assessment according to the WHO, RECIST 1.1, EORTC, and PERCIST criteria in patients diagnosed with malignant solid tumors and who had received cytotoxic chemotherapy to establish the strength of agreement between each criterion. MATERIALS AND METHODS Sixty patients with malignant solid tumors were included in this retrospective study. The baseline and the sequential follow-up fluorine-18-fluorodeoxyglucose PET/computed tomography (CT) of each patient were evaluated according to the WHO, RECIST 1.1, EORTC, and PERCIST criteria. PET/CT images were used for both metabolic and anatomic evaluation. The concurrent diagnostic CT and MRI images (performed within 1 week of PET/CT) were also utilized when needed. The results were compared using the κ-statistics. RESULTS The response and progression rates according to the WHO criteria were 37 and 38%, respectively. The same ratios were also found for RECIST 1.1 (κ=1). The response and progression rates according to the EORTC criteria were 47 and 40%, respectively. When PERCIST criteria were used, one patient with progressive disease was upgraded to stable disease (κ=0.976). As we found the same results with WHO and RECIST 1.1 criteria, we used WHO criteria to compare the anatomic and metabolic criteria. When we compared the WHO and EORTC criteria, there was an agreement in 80% of the patients (κ=0.711). With WHO and PERCIST criteria, there was an agreement in 81.6% of the patients (κ=0.736). CONCLUSION Significant agreement was detected when the WHO, RECIST 1.1, EORTC, and PERCIST criteria were compared both within as well as between each other.
Collapse
|
22
|
Avril S, Muzic RF, Plecha D, Traughber BJ, Vinayak S, Avril N. ¹⁸F-FDG PET/CT for Monitoring of Treatment Response in Breast Cancer. J Nucl Med 2016; 57 Suppl 1:34S-9S. [PMID: 26834099 DOI: 10.2967/jnumed.115.157875] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Changes in tumor metabolic activity have been shown to be an early indicator of treatment effectiveness for breast cancer, mainly in the neoadjuvant setting. The histopathologic response at the completion of chemotherapy has been used as the reference standard for assessment of the accuracy of (18)F-FDG PET in predicting a response during systemic treatment. Although a pathologic complete response (pCR) remains an important positive prognostic factor for an individual patient, a recent metaanalysis could validate pCR as a surrogate marker for patient outcomes only in aggressive breast cancer subtypes. For establishment of the clinical application of metabolic treatment response studies, larger series of specific breast cancer subtypes-including hormone receptor-positive, human epidermal growth factor receptor 2-positive, and triple-negative breast cancers-are necessary. In addition, thresholds for relative changes in (18)F-FDG uptake to distinguish between responding and nonresponding tumors need to be validated for different systemic treatment approaches, with progression-free survival and overall survival as references. A PET-based treatment stratification is applicable clinically only if valid alternative therapies are available. Of note, patients who do not achieve a pCR might still benefit from neoadjuvant therapy enabling breast-conserving surgery. In the metastatic setting, residual tumor metabolic activity after the initiation of systemic therapy is an indicator of active disease, whereas a complete resolution of metabolic activity is predictive of a successful treatment response.
Collapse
Affiliation(s)
- Stefanie Avril
- Department of Pathology, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, Ohio
| | - Raymond F Muzic
- Department of Radiology, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, Ohio
| | - Donna Plecha
- Department of Radiology, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, Ohio
| | - Bryan J Traughber
- Department of Radiation Oncology, Seidman Cancer Center, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, Ohio; and
| | - Shaveta Vinayak
- Department of Medicine (Hematology and Oncology), Seidman Cancer Center, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, Ohio
| | - Norbert Avril
- Department of Radiology, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
23
|
Zaidi H, Thompson C. Evolution and Developments in Instrumentation for Positron Emission Mammography. PET Clin 2016; 4:317-27. [PMID: 27157301 DOI: 10.1016/j.cpet.2009.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Molecular imaging using high-resolution PET instrumentation is now playing a pivotal role in basic and clinical research. The development of optimized detection geometries combined with high-performance detector technologies and compact designs of PET tomographs have become the goal of active research groups in academic and corporate settings. Significant progress has been achieved in the design of commercial PET instrumentation in the last decade allowing a spatial resolution of about 4 to 6 mm to be reached for whole-body imaging, about 2.4 mm for PET cameras dedicated for brain imaging, and submillimeter resolution for female breast, prostate, and small-animal imaging. In particular, significant progress has been made in the design of dedicated positron emission mammography (PEM) units. The initial concept suggested in 1993 consisted of placing 2 planar detectors capable of detecting the 511-keV annihilation photons in a conventional mammography unit. Since that time, many different design paths have been pursued and it will be interesting to see which technologies become the most successful in the future. This paper discusses recent advances in PEM instrumentation and the advantages and challenges of dedicated standalone and dual-modality imaging systems. Future opportunities and the challenges facing the adoption of PEM imaging instrumentation and its role in clinical and research settings are also addressed.
Collapse
Affiliation(s)
- Habib Zaidi
- Division of Nuclear Medicine, Geneva University Hospital, CH-1211 Geneva, Switzerland
| | - Christopher Thompson
- Department of Medical Physics, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| |
Collapse
|
24
|
Al-Muqbel KM, Yaghan RJ. Effectiveness of 18F-FDG-PET/CT vs Bone Scintigraphy in Treatment Response Assessment of Bone Metastases in Breast Cancer. Medicine (Baltimore) 2016; 95:e3753. [PMID: 27227942 PMCID: PMC4902366 DOI: 10.1097/md.0000000000003753] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The aim of the study was to examine the effectiveness of fluorine-18 fluoro-2-deoxy-D-glucose positron emission tomography/computed tomography (18F-FDG PET/CT) versus bone scintigraphy (BS) in treatment response assessment of bone metastases in breast cancer.The medical records of breast cancer patients with metastatic bone disease were reviewed retrospectively in our hospital from the period of January 2003 until April 2014. We included in our study patients evaluated by BS and/or 18F-FDG-PET/CT. Group 1 included patients who underwent pre- and post-treatment BS. Group 2 included patients who underwent pre- and post-treatment 18F-FDG-PET/CT scans. Group 3 included patients who underwent pretreatment BS and post-treatment both modalities. Functional and structural bone changes were monitored on pre- and post-treatment scans.Group 1 included 71 patients, average age of 49.5 y (range 28-73 y). Post-treatment results were as follows: 34% stable disease, 43% progressed disease, 19% improved disease, 3% resolved disease, and 2% relapsed disease. Group 2 included 32 patients, average age 53.2 y (ranges between 37 and 78 y). Post-treatment results were as follows: 3% stable disease, 15% progressed disease, 15% improved disease, 53% resolved disease, and 14% relapsed disease. After treatment, the total symptomatic/imaging concordance rate was 51% in BS and 83% in 18F-FDG-PET/CT. Structurally, most patients with newly diagnosed metastatic bone disease had predominantly osteolytic lesions, which became mixed or osteoblastic after treatment as noted on CT images of responders. Group 3 included 8 patients, average age 48.9 y (ranges 32-64 y). Five patients had stable disease according to BS. 18F-FDG-PET/CT was concordant in 3/5 patients and discordant in 2/5 patients. Three patients had progressed disease on BS with concordant findings on 18F-FDG-PET/CT.18F-FDG-PET/CT was found a powerful tool in treatment response assessment of bone metastases in breast cancer and consistent with clinical status of the patients as it reflects tumor activity. BS is insufficient for response assessment of bone metastases as it reflects osteoblastic reaction of the bone against metastatic disease which increases as the disease responds to treatment.
Collapse
Affiliation(s)
- Kusai M Al-Muqbel
- From the Department of Radiology and Nuclear Medicine and the Department of Surgery, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | | |
Collapse
|
25
|
Jacobs MA, Wolff AC, Macura KJ, Stearns V, Ouwerkerk R, El Khouli R, Bluemke DA, Wahl R. Multiparametric and Multimodality Functional Radiological Imaging for Breast Cancer Diagnosis and Early Treatment Response Assessment. J Natl Cancer Inst Monogr 2016; 2015:40-6. [PMID: 26063885 DOI: 10.1093/jncimonographs/lgv014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Breast cancer is the second leading cause of cancer death among US women, and the chance of a woman developing breast cancer sometime during her lifetime is one in eight. Early detection and diagnosis to allow appropriate locoregional and systemic treatment are key to improve the odds of surviving its diagnosis. Emerging data also suggest that different breast cancer subtypes (phenotypes) may respond differently to available adjuvant therapies. There is a growing understanding that not all patients benefit equally from systemic therapies, and therapeutic approaches are being increasingly personalized based on predictive biomarkers of clinical benefit. Optimal use of established and novel radiological imaging methods, such as magnetic resonance imaging and positron emission tomography, which have different biophysical mechanisms can simultaneously identify key functional parameters. These methods provide unique multiparametric radiological signatures of breast cancer, that will improve the accuracy of early diagnosis, help select appropriate therapies for early stage disease, and allow early assessment of therapeutic benefit.
Collapse
Affiliation(s)
- Michael A Jacobs
- The Russell H. Morgan Department of Radiology and Radiological Science (MAJ, KJM, RO, REK, DAB, RW), Sidney Kimmel Comprehensive Cancer Center (MAJ, ACW, KJM, VS, RW), and Department of Oncology (ACW, VS), The Johns Hopkins University School of Medicine, Baltimore, MD; National Institute of Diabetes and Digestive and Kidney Diseases (not affialted, RO) and Radiology and Imaging Sciences, National Institutes of Health Clinical Center (DAB), Bethesda, MD.
| | - Antonio C Wolff
- The Russell H. Morgan Department of Radiology and Radiological Science (MAJ, KJM, RO, REK, DAB, RW), Sidney Kimmel Comprehensive Cancer Center (MAJ, ACW, KJM, VS, RW), and Department of Oncology (ACW, VS), The Johns Hopkins University School of Medicine, Baltimore, MD; National Institute of Diabetes and Digestive and Kidney Diseases (not affialted, RO) and Radiology and Imaging Sciences, National Institutes of Health Clinical Center (DAB), Bethesda, MD
| | - Katarzyna J Macura
- The Russell H. Morgan Department of Radiology and Radiological Science (MAJ, KJM, RO, REK, DAB, RW), Sidney Kimmel Comprehensive Cancer Center (MAJ, ACW, KJM, VS, RW), and Department of Oncology (ACW, VS), The Johns Hopkins University School of Medicine, Baltimore, MD; National Institute of Diabetes and Digestive and Kidney Diseases (not affialted, RO) and Radiology and Imaging Sciences, National Institutes of Health Clinical Center (DAB), Bethesda, MD
| | - Vered Stearns
- The Russell H. Morgan Department of Radiology and Radiological Science (MAJ, KJM, RO, REK, DAB, RW), Sidney Kimmel Comprehensive Cancer Center (MAJ, ACW, KJM, VS, RW), and Department of Oncology (ACW, VS), The Johns Hopkins University School of Medicine, Baltimore, MD; National Institute of Diabetes and Digestive and Kidney Diseases (not affialted, RO) and Radiology and Imaging Sciences, National Institutes of Health Clinical Center (DAB), Bethesda, MD
| | - Ronald Ouwerkerk
- The Russell H. Morgan Department of Radiology and Radiological Science (MAJ, KJM, RO, REK, DAB, RW), Sidney Kimmel Comprehensive Cancer Center (MAJ, ACW, KJM, VS, RW), and Department of Oncology (ACW, VS), The Johns Hopkins University School of Medicine, Baltimore, MD; National Institute of Diabetes and Digestive and Kidney Diseases (not affialted, RO) and Radiology and Imaging Sciences, National Institutes of Health Clinical Center (DAB), Bethesda, MD
| | - Riham El Khouli
- The Russell H. Morgan Department of Radiology and Radiological Science (MAJ, KJM, RO, REK, DAB, RW), Sidney Kimmel Comprehensive Cancer Center (MAJ, ACW, KJM, VS, RW), and Department of Oncology (ACW, VS), The Johns Hopkins University School of Medicine, Baltimore, MD; National Institute of Diabetes and Digestive and Kidney Diseases (not affialted, RO) and Radiology and Imaging Sciences, National Institutes of Health Clinical Center (DAB), Bethesda, MD
| | - David A Bluemke
- The Russell H. Morgan Department of Radiology and Radiological Science (MAJ, KJM, RO, REK, DAB, RW), Sidney Kimmel Comprehensive Cancer Center (MAJ, ACW, KJM, VS, RW), and Department of Oncology (ACW, VS), The Johns Hopkins University School of Medicine, Baltimore, MD; National Institute of Diabetes and Digestive and Kidney Diseases (not affialted, RO) and Radiology and Imaging Sciences, National Institutes of Health Clinical Center (DAB), Bethesda, MD
| | - Richard Wahl
- The Russell H. Morgan Department of Radiology and Radiological Science (MAJ, KJM, RO, REK, DAB, RW), Sidney Kimmel Comprehensive Cancer Center (MAJ, ACW, KJM, VS, RW), and Department of Oncology (ACW, VS), The Johns Hopkins University School of Medicine, Baltimore, MD; National Institute of Diabetes and Digestive and Kidney Diseases (not affialted, RO) and Radiology and Imaging Sciences, National Institutes of Health Clinical Center (DAB), Bethesda, MD
| |
Collapse
|
26
|
Semiglazov V. RECIST for Response (Clinical and Imaging) in Neoadjuvant Clinical Trials in Operable Breast Cancer. J Natl Cancer Inst Monogr 2016; 2015:21-3. [PMID: 26063880 DOI: 10.1093/jncimonographs/lgv021] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Although approximately 70% of breast cancer patients demonstrate a clinical response on neoadjuvant systemic therapy on physical examination or on anatomic radiographic imaging, only 3%-40% achieve a pathologic complete response (pCR). Magnetic resonance imaging (MRI) is superior to physical examination, ultrasound, and mammography in response evaluation during neoadjuvant systemic therapy. The accuracy of breast MRI to predict pCR has a moderate sensitivity, but high specificity. The accuracy of anatomic imaging to assess residual disease and predict pCR depended on anatomic radiographic imaging cancer subtypes. Response monitoring using breast is accurate in triple-negative or HER2-positive tumors. It was inaccurate in estrogen receptor-positive/HER2-negative subtype. Another approach currently under investigation is dynamic contrast-enhanced MRI and diffusion weighted-imaging, (18)F-fluorodeoxyglucose positron emission tomography, fluorodeoxyglucose positron emission tomography/computed tomography.
Collapse
|
27
|
Lee HW, Lee HM, Choi SE, Yoo H, Ahn SG, Lee MK, Jeong J, Jung WH. The Prognostic Impact of Early Change in 18F-FDG PET SUV After Neoadjuvant Chemotherapy in Patients with Locally Advanced Breast Cancer. J Nucl Med 2016; 57:1183-8. [DOI: 10.2967/jnumed.115.166322] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 03/08/2016] [Indexed: 01/28/2023] Open
|
28
|
Reporter nanoparticle that monitors its anticancer efficacy in real time. Proc Natl Acad Sci U S A 2016; 113:E2104-13. [PMID: 27036008 DOI: 10.1073/pnas.1603455113] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The ability to monitor the efficacy of an anticancer treatment in real time can have a critical effect on the outcome. Currently, clinical readouts of efficacy rely on indirect or anatomic measurements, which occur over prolonged time scales postchemotherapy or postimmunotherapy and may not be concordant with the actual effect. Here we describe the biology-inspired engineering of a simple 2-in-1 reporter nanoparticle that not only delivers a cytotoxic or an immunotherapy payload to the tumor but also reports back on the efficacy in real time. The reporter nanoparticles are engineered from a novel two-staged stimuli-responsive polymeric material with an optimal ratio of an enzyme-cleavable drug or immunotherapy (effector elements) and a drug function-activatable reporter element. The spatiotemporally constrained delivery of the effector and the reporter elements in a single nanoparticle produces maximum signal enhancement due to the availability of the reporter element in the same cell as the drug, thereby effectively capturing the temporal apoptosis process. Using chemotherapy-sensitive and chemotherapy-resistant tumors in vivo, we show that the reporter nanoparticles can provide a real-time noninvasive readout of tumor response to chemotherapy. The reporter nanoparticle can also monitor the efficacy of immune checkpoint inhibition in melanoma. The self-reporting capability, for the first time to our knowledge, captures an anticancer nanoparticle in action in vivo.
Collapse
|
29
|
Tőkés T, Kajáry K, Szentmártoni G, Lengyel Z, Györke T, Torgyík L, Somlai K, Tőkés AM, Kulka J, Dank M. Predictive and prognostic value of FDG-PET/CT imaging and different response evaluation criteria after primary systemic therapy of breast cancer. Breast Cancer 2016; 24:137-146. [PMID: 26979062 DOI: 10.1007/s12282-016-0685-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 03/04/2016] [Indexed: 12/27/2022]
Abstract
OBJECTIVES (1) To predict pathological complete remission (pCR) and survival after primary systemic therapy (PST) in patients diagnosed with breast cancer by using two different PET/CT based scores: a simplified PERCIST-based PET/CT score (Method 1) and a combined PET/CT score supplemented with the morphological results of the RECIST system (Method 2) and (2) to assess the effect of different breast carcinoma subtypes on tumor response and its evaluation. METHODS Eighty-eight patients were enrolled in the study who underwent PET/CT imaging before and after PST. PET/CTs were evaluated by changes in maximum Standardized Uptake Value (SUVmax) and tumor size. Method 1 and 2 were applied to predict pathological complete remission (pCR). Kaplan-Meier analyses for survival were performed. Classification into biological subtypes was performed based on the pre-therapeutic tumor characteristics. RESULTS A total of 30/88 patients showed pCR (34.1 %). Comparing pCR/non-pCR patient groups, significant differences were detected by changes in SUVmax (p < 0.001) and tumor size (p < 0.001) regarding the primary breast lesions. To predict pCR, Method 2 had higher sensitivity (72.4 % vs. 44.8 %) and negative predictive value (57.9 % vs. 45.8 %) with lower false negativity rate (16 vs. 32) than Method 1. pCR rate was higher in Her2-positive and triple negative tumors. Despite the significant differences detected between the biological subtypes regarding changes in primary tumor SUVmax (p = 0.007) and size (p = 0.015), the subtypes only had significant impact on response evaluation with Method 2 and not with Method 1. In our study, neither clinical nor pathological CR were predictors of longer progression-free survival. CONCLUSIONS Our results suggest that combined PET/CT criteria are more predictive of pCR. The effect of biological subtypes is significant on pCR rate as well as on the changes in FDG-uptake and morphological tumor response. Response evaluation with combined criteria was also able to reflect the differences between the biological behavior of breast tumor subtypes.
Collapse
Affiliation(s)
- Tímea Tőkés
- Oncological Division, 1st Department of Internal Medicine, Semmelweis University, Tömő utca 25-29, 4th floor, Budapest, 1083, Hungary
| | - Kornélia Kajáry
- Pozitron PET/CT Center, Hunyadi J. út 9, Budapest, 1117, Hungary
| | - Gyöngyvér Szentmártoni
- Oncological Division, 1st Department of Internal Medicine, Semmelweis University, Tömő utca 25-29, 4th floor, Budapest, 1083, Hungary
| | - Zsolt Lengyel
- Pozitron PET/CT Center, Hunyadi J. út 9, Budapest, 1117, Hungary
| | - Tamás Györke
- Department of Nuclear Medicine, Semmelweis University, Üllői út 78/A, Budapest, 1082, Hungary.,Scanomed Ltd, Laky Adolf utca 44, Budapest, 1145, Hungary
| | - László Torgyík
- Oncological Division, 1st Department of Internal Medicine, Semmelweis University, Tömő utca 25-29, 4th floor, Budapest, 1083, Hungary
| | - Krisztián Somlai
- Surgical Division of the St Margaret Hospital, Bécsi út 132, Budapest, 1032, Hungary
| | - Anna-Mária Tőkés
- MTA-SE Tumor Progression Research Group, 2nd Department of Pathology, Üllői út 93, Budapest, 1091, Hungary.,2nd Department of Pathology, Semmelweis University, Üllői út 93, Budapest, 1091, Hungary
| | - Janina Kulka
- MTA-SE Tumor Progression Research Group, 2nd Department of Pathology, Üllői út 93, Budapest, 1091, Hungary.,2nd Department of Pathology, Semmelweis University, Üllői út 93, Budapest, 1091, Hungary
| | - Magdolna Dank
- Oncological Division, 1st Department of Internal Medicine, Semmelweis University, Tömő utca 25-29, 4th floor, Budapest, 1083, Hungary.
| |
Collapse
|
30
|
Dewalle-Vignion AS, Betrouni N, Baillet C, Vermandel M. Is STAPLE algorithm confident to assess segmentation methods in PET imaging? Phys Med Biol 2015; 60:9473-91. [PMID: 26584044 DOI: 10.1088/0031-9155/60/24/9473] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Accurate tumor segmentation in [18F]-fluorodeoxyglucose positron emission tomography is crucial for tumor response assessment and target volume definition in radiation therapy. Evaluation of segmentation methods from clinical data without ground truth is usually based on physicians' manual delineations. In this context, the simultaneous truth and performance level estimation (STAPLE) algorithm could be useful to manage the multi-observers variability. In this paper, we evaluated how this algorithm could accurately estimate the ground truth in PET imaging. Complete evaluation study using different criteria was performed on simulated data. The STAPLE algorithm was applied to manual and automatic segmentation results. A specific configuration of the implementation provided by the Computational Radiology Laboratory was used. Consensus obtained by the STAPLE algorithm from manual delineations appeared to be more accurate than manual delineations themselves (80% of overlap). An improvement of the accuracy was also observed when applying the STAPLE algorithm to automatic segmentations results. The STAPLE algorithm, with the configuration used in this paper, is more appropriate than manual delineations alone or automatic segmentations results alone to estimate the ground truth in PET imaging. Therefore, it might be preferred to assess the accuracy of tumor segmentation methods in PET imaging.
Collapse
Affiliation(s)
- Anne-Sophie Dewalle-Vignion
- Université Lille, Inserm, CHU Lille, U1189-ONCO-THAI-Image Assisted Laser Therapy for Oncology, F-59000 Lille, France
| | | | | | | |
Collapse
|
31
|
Michaels AY, Keraliya AR, Tirumani SH, Shinagare AB, Ramaiya NH. Systemic treatment in breast cancer: a primer for radiologists. Insights Imaging 2015; 7:131-44. [PMID: 26567115 PMCID: PMC4729711 DOI: 10.1007/s13244-015-0447-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 09/24/2015] [Accepted: 10/21/2015] [Indexed: 12/22/2022] Open
Abstract
Abstract Cytotoxic chemotherapy, hormonal therapy and molecular targeted therapy are the three major classes of drugs used to treat breast cancer. Imaging modalities such as computed tomography (CT), magnetic resonance imaging (MRI), 18F-FDG positron emission tomography (PET)/CT and bone scintigraphy each have a distinct role in monitoring response and detecting drug toxicities associated with these treatments. The purpose of this article is to elucidate the various systemic therapies used in breast cancer, with an emphasis on the role of imaging in assessing treatment response and detecting treatment-related toxicities. Teaching Points • Cytotoxic chemotherapy is often used in combination with HER2-targeted and endocrine therapies. • Endocrine and HER2-targeted therapies are recommended in hormone-receptor- and HER2-positive cases. • CT is the workhorse for assessment of treatment response in breast cancer metastases. • Alternate treatment response criteria can help in interpreting pseudoprogression in metastasis. • Unique toxicities are associated with cytotoxic chemotherapy and with endocrine and HER2-targeted therapies.
Collapse
Affiliation(s)
- Aya Y Michaels
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Abhishek R Keraliya
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.,Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Sree Harsha Tirumani
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA. .,Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA.
| | - Atul B Shinagare
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.,Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Nikhil H Ramaiya
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.,Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
| |
Collapse
|
32
|
Maus J, Schramm G, Hofheinz F, Oehme L, Lougovski A, Petr J, Platzek I, Beuthien-Baumann B, Steinbach J, Kotzerke J, van den Hoff J. Evaluation of in vivo quantification accuracy of the Ingenuity-TF PET/MR. Med Phys 2015; 42:5773-81. [PMID: 26429251 DOI: 10.1118/1.4929936] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
PURPOSE The quantitative accuracy of standardized uptake values (SUVs) and tracer kinetic uptake parameters in patient investigations strongly depends on accurate determination of regional activity concentrations in positron emission tomography (PET) data. This determination rests on the assumption that the given scanner calibration is valid in vivo. In a previous study, we introduced a method to test this assumption. This method allows to identify discrepancies in quantitative accuracy in vivo by comparison of activity concentrations of urine samples measured in a well-counter with activity concentrations extracted from PET images of the bladder. In the present study, we have applied this method to the Philips Ingenuity-TF PET/MR since at the present stage, absolute quantitative accuracy of combined PET/MR systems is still under investigation. METHODS Twenty one clinical whole-body F18-FDG scans were included in this study. The bladder region was imaged as the last bed position and urine samples were collected afterward. PET images were reconstructed including MR-based attenuation correction with and without truncation compensation and 3D regions-of-interest (ROIs) of the bladder were delineated by three observers. To exclude partial volume effects, ROIs were concentrically shrunk by 8-10 mm. Then, activity concentrations were determined in the PET images for the bladder and for the urine by measuring the samples in a calibrated well-counter. In addition, linearity measurements of SUV vs singles rate and measurements of the stability of the coincidence rate of "true" events of the PET/MR system were performed over a period of 4 months. RESULTS The measured in vivo activity concentrations were significantly lower in PET/MR than in the well-counter with a ratio of the former to the latter of 0.756 ± 0.060 (mean ± std. dev.), a range of 0.604-0.858, and a P value of 3.9 ⋅ 10(-14). While the stability measurements of the coincidence rate of "true" events showed no relevant deviation over time, the linearity scans revealed a systematic error of 8%-11% (avg. 9%) for the range of singles rates present in the bladder scans. After correcting for this systematic bias caused by shortcomings of the manufacturers calibration procedure, the PET to well-counter ratio increased to 0.832 ± 0.064 (0.668 -0.941), P = 1.1 ⋅ 10(-10). After compensating for truncation of the upper extremities in the MR-based attenuation maps, the ratio further improved to 0.871 ± 0.069 (0.693-0.992), P = 3.9 ⋅ 10(-8). CONCLUSIONS Our results show that the Philips PET/MR underestimates activity concentrations in the bladder by 17%, which is 7 percentage points (pp.) larger than in the previously investigated PET and PET/CT systems. We attribute this increased underestimation to remaining limitations of the MR-based attenuation correction. Our results suggest that only a 2 pp. larger underestimation of activity concentrations compared to PET/CT can be observed if compensation of attenuation truncation of the upper extremities is applied. Thus, quantification accuracy of the Philips Ingenuity-TF PET/MR can be considered acceptable for clinical purposes given the ±10% error margin in the EANM guidelines. The comparison of PET images from the bladder region with urine samples has proven a useful method. It might be interesting for evaluation and comparison of the in vivo quantitative accuracy of PET, PET/CT, and especially PET/MR systems from different manufacturers or in multicenter trials.
Collapse
Affiliation(s)
- Jens Maus
- PET Center, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, P.O. Box 510119, 01314 Dresden, Germany
| | - Georg Schramm
- PET Center, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, P.O. Box 510119, 01314 Dresden, Germany
| | - Frank Hofheinz
- PET Center, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, P.O. Box 510119, 01314 Dresden, Germany
| | - Liane Oehme
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Alexandr Lougovski
- PET Center, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, P.O. Box 510119, 01314 Dresden, Germany
| | - Jan Petr
- PET Center, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, P.O. Box 510119, 01314 Dresden, Germany
| | - Ivan Platzek
- Department of Radiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Bettina Beuthien-Baumann
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Jörg Steinbach
- PET Center, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, P.O. Box 510119, 01314 Dresden, Germany
| | - Jörg Kotzerke
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Jörg van den Hoff
- PET Center, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, P.O. Box 510119, 01314 Dresden, Germany and Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| |
Collapse
|
33
|
Kang WJ, Song EH, Park JY, Park YJ, Cho A, Song HT. 18F-fluoride PET imaging in a nude rat model of bone metastasis from breast cancer: Comparison with 18F-FDG and bioluminescence imaging. Nucl Med Biol 2015; 42:728-33. [DOI: 10.1016/j.nucmedbio.2015.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 05/01/2015] [Accepted: 05/20/2015] [Indexed: 12/15/2022]
|
34
|
He H, Cai C, Charnsangavej C, Theriault RL, Green M, Quraishi MA, Yang WT. Contrast-Enhanced Computed Tomography Evaluation of Hepatic Metastases in Breast Cancer Patients Before and After Cytotoxic Chemotherapy or Targeted Therapy. Can Assoc Radiol J 2015; 66:356-62. [PMID: 26165625 DOI: 10.1016/j.carj.2015.03.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 02/08/2015] [Accepted: 03/03/2015] [Indexed: 01/13/2023] Open
Abstract
PURPOSE To evaluate change in size vs computed tomography (CT) density of hepatic metastases in breast cancer patients before and after cytotoxic chemotherapy or targeted therapy. METHODS A database search in a single institution identified 48 breast cancer patients who had hepatic metastases treated with either cytotoxic chemotherapy alone or targeted therapy alone, and who had contrast-enhanced CT (CECT) scans of the abdomen at baseline and within 4 months of initiation of therapy in the past 10 years. Two radiologists retrospectively evaluated CT scans and identified up to 2 index lesions in each patient. The size (centimeters) of each lesion was measured according to Response Evaluation Criteria in Solid Tumors (RECIST) criteria, and CT density (Hounsfield units) was measured by drawing a region of interest around the margin of the entire lesion. The percent change in sum of lesion size and mean CT density on pre- and post-treatment scans was computed for each patient; results were compared within each treatment group. RESULTS Thirty-nine patients with 68 lesions received cytotoxic chemotherapy only; 9 patients with 15 lesions received targeted therapy only. The mean percent changes in sum of lesion size and mean CT density were statistically significant within the cytotoxic chemotherapy group before and after treatment, but not significant in the targeted therapy group. The patients in the targeted therapy group tend to have better 2-year survival. The patients who survived at 2 years tend to have more decrease in tumour size in the cytotoxic chemotherapy group. CONCLUSION Cytotoxic chemotherapy produced significant mean percent decrease in tumour size and mean CT density of hepatic metastases from breast cancer before and after treatment, whereas targeted therapy did not. Nonetheless, there is a trend that the patients in the targeted therapy group had better 2-year survival rate. This suggests that RECIST is potentially inadequate in evaluating tumour response in breast cancer liver metastases treated with targeted therapy alone, calling for an alternative marker for response evaluation in this subset of patients.
Collapse
Affiliation(s)
- Hongying He
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Diagnostic and Interventional Imaging, The University of Texas Health Science Center at Houston, Houston, Texas, USA.
| | - Chunyan Cai
- Division of Clinical and Translational Sciences, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Chusilp Charnsangavej
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Richard L Theriault
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Marjorie Green
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mohammad A Quraishi
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wei T Yang
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
35
|
Tőkés T, Szentmártoni G, Torgyík L, Kajáry K, Lengyel Z, Györke T, Molnár BÁ, Tőkés AM, Kulka J, Dank M. Response evaluation after primary systemic therapy of Her2 positive breast cancer – an observational cross-sectional study. Croat Med J 2015; 56:128-38. [PMID: 25891872 PMCID: PMC4410165 DOI: 10.3325/cmj.2015.56.128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim To evaluate (I) trastuzumab-containing primary systemic therapy (PST) in human epidermal growth factor receptor 2 (Her2) overexpressing breast carcinomas; (II) compare the patients who achieved and those who did not achieve pathological complete remission (pCR), and (III) analyze the accuracy of different clinical-imaging modalities in tumor response monitoring. Methods 188 patients who received PST between 2008 and 2014 were reviewed and 43 Her2 overexpressing breast cancer patients (28 Luminal B/Her2-positive and 15 Her2-positive) were enrolled. 26 patients received mostly taxane-based PST without trastuzumab (Group 1) and 17 patients received trastuzumab-containing PST (Group 2). We compared the concordance between pCR and complete remission (CR) defined by breast-ultrasound, CR defined by standard 18F-fluoro-deoxy-glucose positron emission tomography and computerized tomography (FDG-PET/CT) criteria (Method 1) and CR defined by a novel, breast cancer specific FDG-PET/CT criteria (Method 2). Sensitivity (sens), specificity (spec), and positive (PPV) and negative predictive values (NPV) were calculated. Results 10 patients (38.5%) in Group 1 and 8 (47%) in Group 2 achieved pCR. pCR was significantly more frequent in Her2-positive than in Luminal B/Her2-positive tumors in both Group 1 (P = 0.043) and Group 2 (P = 0.029). PET/CT evaluated by the breast cancer specific criteria (PET/CT Method 2) differentiated pCR from non-pCR more accurately in both groups (Group 1: sens = 77.8%, spec = 100%, PPV = 100%, NPV = 71.4%; Group 2: sens = 87.5%, spec = 62.5%, PPV = 70%, NPV = 83.3%) than standard PET/CT criteria (Method 1) (Group 1: sens = 22.2% spec = 100% PPV = 100% NPV = 41.7%; in Group 2: sens = 37.5%, spec = 87.5%, PPV = 75% NPV = 58.3%) or breast ultrasound (Group 1, sens = 83.3% spec = 25% PPV = 62.5% NPV = 50%; Group 2, sens = 100% spec = 12.5% PPV = 41.6% NPV = 100%). Conclusion The benefit of targeted treatment with trastuzumab-containing PST in Her2 overexpressing breast cancer was defined in terms of pCR rate. Luminal B/Her2-positive subtype needs further subdivision to identify patients who would benefit from PST. Combined evaluation of tumor response by our novel, breast cancer specific FDG-PET/CT criteria accurately differentiated pCR from non-pCR patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Magdolna Dank
- Magdolna Dank, Semmelweis University, 1st Dept. of Internal Medicine, Oncological Division, Tömő street 25-29. 4th floor, Budapest, Hungary, H- 1083.,
| |
Collapse
|
36
|
Humbert O, Cochet A, Coudert B, Berriolo-Riedinger A, Kanoun S, Brunotte F, Fumoleau P. Role of positron emission tomography for the monitoring of response to therapy in breast cancer. Oncologist 2015; 20:94-104. [PMID: 25561512 DOI: 10.1634/theoncologist.2014-0342] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
This review considers the potential utility of positron emission tomography (PET) tracers in the setting of response monitoring in breast cancer, with a special emphasis on glucose metabolic changes assessed with (18)F-fluorodeoxyglucose (FDG). In the neoadjuvant setting of breast cancer, the metabolic response can predict the final complete pathologic response after the first cycles of chemotherapy. Because tumor metabolic behavior highly depends on cancer subtype, studies are ongoing to define the optimal metabolic criteria of tumor response in each subtype. The recent multicentric randomized AVATAXHER trial has suggested, in the human epidermal growth factor 2-positive subtype, a clinical benefit of early tailoring the neoadjuvant treatment in women with poor metabolic response after the first course of treatment. In the bone-dominant metastatic setting, there is increasing clinical evidence that FDG-PET/computed tomography (CT) is the most accurate imaging modality for assessment of the tumor response to treatment when both metabolic information and morphologic information are considered. Nevertheless, there is a need to define standardized metabolic criteria of response, including the heterogeneity of response among metastases, and to evaluate the costs and health outcome of FDG-PET/CT compared with conventional imaging. New non-FDG radiotracers highlighting specific molecular hallmarks of breast cancer cells have recently emerged in preclinical and clinical studies. These biomarkers can take into account the heterogeneity of tumor biology in metastatic lesions. They may provide valuable clinical information for physicians to select and monitor the effectiveness of novel therapeutics targeting the same molecular pathways of breast tumor.
Collapse
Affiliation(s)
- Olivier Humbert
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| | - Alexandre Cochet
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| | - Bruno Coudert
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| | - Alina Berriolo-Riedinger
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| | - Salim Kanoun
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| | - François Brunotte
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| | - Pierre Fumoleau
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| |
Collapse
|
37
|
|
38
|
Gallamini A, Zwarthoed C, Borra A. Positron Emission Tomography (PET) in Oncology. Cancers (Basel) 2014; 6:1821-89. [PMID: 25268160 PMCID: PMC4276948 DOI: 10.3390/cancers6041821] [Citation(s) in RCA: 217] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 07/25/2014] [Accepted: 08/07/2014] [Indexed: 02/07/2023] Open
Abstract
Since its introduction in the early nineties as a promising functional imaging technique in the management of neoplastic disorders, FDG-PET, and subsequently FDG-PET/CT, has become a cornerstone in several oncologic procedures such as tumor staging and restaging, treatment efficacy assessment during or after treatment end and radiotherapy planning. Moreover, the continuous technological progress of image generation and the introduction of sophisticated software to use PET scan as a biomarker paved the way to calculate new prognostic markers such as the metabolic tumor volume (MTV) and the total amount of tumor glycolysis (TLG). FDG-PET/CT proved more sensitive than contrast-enhanced CT scan in staging of several type of lymphoma or in detecting widespread tumor dissemination in several solid cancers, such as breast, lung, colon, ovary and head and neck carcinoma. As a consequence the stage of patients was upgraded, with a change of treatment in 10%-15% of them. One of the most evident advantages of FDG-PET was its ability to detect, very early during treatment, significant changes in glucose metabolism or even complete shutoff of the neoplastic cell metabolism as a surrogate of tumor chemosensitivity assessment. This could enable clinicians to detect much earlier the effectiveness of a given antineoplastic treatment, as compared to the traditional radiological detection of tumor shrinkage, which usually takes time and occurs much later.
Collapse
Affiliation(s)
- Andrea Gallamini
- Department of Research and Medical Innovation, Antoine Lacassagne Cancer Center, Nice University, Nice Cedex 2-06189 Nice, France.
| | - Colette Zwarthoed
- Department of Nuclear Medicine, Antoine Lacassagne Cancer Center, Nice University, Nice Cedex 2-06189 Nice, France.
| | - Anna Borra
- Hematology Department S. Croce Hospital, Via M. Coppino 26, Cuneo 12100, Italy.
| |
Collapse
|
39
|
Sher A, Valls L, Muzic RF, Plecha D, Avril N. Whole-body positron emission tomography-magnetic resonance in breast cancer. Semin Roentgenol 2014; 49:313-20. [PMID: 25498228 DOI: 10.1053/j.ro.2014.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Andrew Sher
- Department of Radiology, University Hospitals Case Medical Center, Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH
| | - Laia Valls
- Department of Radiology, University Hospitals Case Medical Center, Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH
| | - Raymond F Muzic
- Department of Radiology, University Hospitals Case Medical Center, Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH
| | - Donna Plecha
- Department of Radiology, University Hospitals Case Medical Center, Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH
| | - Norbert Avril
- Department of Radiology, University Hospitals Case Medical Center, Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH.
| |
Collapse
|
40
|
Groheux D, Hindié E, Marty M, Espié M, Rubello D, Vercellino L, Bousquet G, Ohnona J, Toubert ME, Merlet P, Misset JL. ¹⁸F-FDG-PET/CT in staging, restaging, and treatment response assessment of male breast cancer. Eur J Radiol 2014; 83:1925-33. [PMID: 24985086 DOI: 10.1016/j.ejrad.2014.05.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 05/27/2014] [Accepted: 05/28/2014] [Indexed: 10/25/2022]
Abstract
PURPOSE Male breast cancer (BC) is a rare disease, with patterns different from those found in women. Most tumors are detected at more advanced stages than in women. The aim of this study was to analyze the performance of [18F]fluorodeoxyglucose positron emission tomography/computed tomography ((18)F-FDG-PET/CT) in staging, restaging, and therapy response assessment. METHODS We performed a systematic analysis in the database of Saint-Louis Hospital to identify male patients with BC referred for PET/CT. (18)F-FDG-PET/CT findings considered suspicious for malignancy were compared to biopsy results, further work-up and/or patient follow-up of at least 6 months. Performances of (18)F-FDG-PET/CT were compared to that of conventional imaging (CI) using the McNemar test. The impact of PET/CT on management was evaluated. RESULTS During 6 consecutive years, among 12,692 (18)F-FDG-PET/CT oncology studies, 30 were performed in 15 men with BC: 7 examinations for initial staging, 11 for restaging, and 12 for response assessment. Tumors profile was ER+ and one had HER2 overexpression. PET/CT sensitivity, specificity, positive predictive value, negative predictive value and accuracy to detect distant metastases were 100%, 67%, 86%, 100% and 89%, respectively. PET/CT was more informative than CI in 40% of studies (p=0.03; 95% confidence interval: 3.26 - 40%). Findings from (18)F-FDG-PET/CT led to modification in the planned treatment in 13/30 cases (43%). CONCLUSION Although all the tumors were ER+, primary lesions and metastases were diagnosed with high sensitivity. (18)F-FDG-PET/CT seems to be a powerful imaging method to perform staging, restaging and treatment response assessment in male patients with BC.
Collapse
Affiliation(s)
- David Groheux
- Department of Nuclear Medicine, Saint-Louis Hospital, Paris, France; Doctoral School of Biology and Biotechnology, University Institute of Hematology, University of Paris VII, Paris, France.
| | - Elif Hindié
- Department of Nuclear Medicine, Haut-Lévêque Hospital, CHU Bordeaux, University Bordeaux-Segalen, Bordeaux, France
| | - Michel Marty
- Breast Diseases Unit and Department of Medical Oncology, Saint-Louis Hospital, Paris, France; Centre for Therapeutic Innovation, Saint-Louis Hospital, Paris, France
| | - Marc Espié
- Breast Diseases Unit and Department of Medical Oncology, Saint-Louis Hospital, Paris, France
| | - Domenico Rubello
- Department of Nuclear Medicine, Santa Maria della Misericordia, Rovigo Hospital, Rovigo, Italy
| | - Laetitia Vercellino
- Department of Nuclear Medicine, Saint-Louis Hospital, Paris, France; Doctoral School of Biology and Biotechnology, University Institute of Hematology, University of Paris VII, Paris, France
| | - Guilhem Bousquet
- Breast Diseases Unit and Department of Medical Oncology, Saint-Louis Hospital, Paris, France; INSERM U728, University Institute of Hematology, University of Paris VII, Paris, France
| | - Jessica Ohnona
- Department of Nuclear Medicine, Saint-Louis Hospital, Paris, France
| | | | - Pascal Merlet
- Department of Nuclear Medicine, Saint-Louis Hospital, Paris, France; Doctoral School of Biology and Biotechnology, University Institute of Hematology, University of Paris VII, Paris, France
| | - Jean-Louis Misset
- Breast Diseases Unit and Department of Medical Oncology, Saint-Louis Hospital, Paris, France
| |
Collapse
|
41
|
Magometschnigg HF, Helbich T, Brader P, Abeyakoon O, Baltzer P, Füger B, Wengert G, Polanec S, Bickel H, Pinker K. Molecular imaging for the characterization of breast tumors. Expert Rev Anticancer Ther 2014; 14:711-22. [DOI: 10.1586/14737140.2014.885383] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
42
|
Predictive value of PET-CT for pathological response in stages II and III breast cancer patients following neoadjuvant chemotherapy with docetaxel. Rev Esp Med Nucl Imagen Mol 2014. [DOI: 10.1016/j.remnie.2013.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
43
|
Andrade W, Lima E, Osório C, do Socorro Maciel M, Baiocchi G, Bitencourt A, Fanelli M, Damascena A, Soares F. Can FDG-PET/CT predict early response to neoadjuvant chemotherapy in breast cancer? Eur J Surg Oncol 2013; 39:1358-63. [DOI: 10.1016/j.ejso.2013.08.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Revised: 08/22/2013] [Accepted: 08/23/2013] [Indexed: 12/30/2022] Open
|
44
|
Paudyal B, Zhang K, Chen CP, Wampole ME, Mehta N, Mitchell EP, Gray BD, Mattis JA, Pak KY, Thakur ML, Wickstrom E. Determining efficacy of breast cancer therapy by PET imaging of HER2 mRNA. Nucl Med Biol 2013; 40:994-9. [PMID: 24074944 DOI: 10.1016/j.nucmedbio.2013.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 07/30/2013] [Accepted: 08/17/2013] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Monitoring the effectiveness of therapy early and accurately continues to be challenging. We hypothesize that determination of Human Epidermal Growth Factor Receptor 2 (HER2) mRNA in malignant breast cancer (BC) cells by positron emission tomography (PET) imaging, before and after treatment, would reflect therapeutic efficacy. METHOD WT4340, a peptide nucleic acid (PNA) 12-mer complementary to HER2 mRNA was synthesized together with -CSKC, a cyclic peptide, which facilitated internalization of the PNA via IGFR expressed on BC cells, and DOTA that chelated Cu-64. Mice (n = 8) with BT474 ER+/HER2+ human BC received doxorubicin (DOX, 1.5mg/kg) i.p. once a week for six weeks. Mice (n = 8) without DOX served as controls. All mice were PET imaged with F-18-FDG and 48 h later with Cu-64-WT4340. PET imaging were performed before and 72 h after each treatment. Standardized uptake values (SUVs) were determined and percent change calculated. Animal body weight (BW) and tumor volume (TV) were measured. RESULTS SUVs for Cu-64-WT4340 after DOX treatment declined by 54% ± 17% after the second dose, 41% ± 15% after the fourth dose, and 29% ± 7% after the sixth dose, compared with 42% ± 22%, 31% ± 18%, and 13% ± 9% (p<0.05) for F-18-FDG. In untreated mice, the corresponding percent SUVs for Cu-64-WT4340 were 145% ± 82%, 165% ± 39%, and 212% ± 105% of pretreatment SUV, compared with 108% ± 28%, 151% ± 8%, and 152% ± 35.5%, (p<0.08) for F-18-FDG. TV in mice after second dose was 114.15% ± 61.83%, compared with 144.7% ± 64.4% for control mice. BW of DOX-treated mice was 103.4% ± 7.6% of pretreatment, vs. 100.1% ± 4.3% for control mice. CONCLUSION Therapeutic efficacy was apparent sooner by molecular PET imaging than by determination of reduction in TV.
Collapse
Affiliation(s)
- Bishnuhari Paudyal
- Department of Radiology, Thomas Jefferson University, 1020 Locust Street, JAH 474, Philadelphia PA 19107, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
García García-Esquinas MA, Arrazola García J, García-Sáenz JA, Furió-Bacete V, Fuentes Ferrer ME, Ortega Candil A, Cabrera Martín MN, Carreras Delgado JL. Predictive value of PET-CT for pathological response in stages II and III breast cancer patients following neoadjuvant chemotherapy with docetaxel. Rev Esp Med Nucl Imagen Mol 2013; 33:14-21. [PMID: 23809513 DOI: 10.1016/j.remn.2013.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Revised: 04/23/2013] [Accepted: 04/25/2013] [Indexed: 10/26/2022]
Abstract
PURPOSE To prospectively study the value of PET-CT with fluorine-18 fluorodeoxyglucose (FDG) to predict neoadjuvant chemotherapy (NAC) response of locoregional disease of stages II and III breast cancer patients. MATERIAL AND METHODS A written informed consent and approval were obtained from the Ethics Committee. PET-CT accuracy in the prediction of pathologic complete response (pCR) after NAC was studied in primary tumors and lymph node metastasis in 43 women (mean age: 50 years: range: 27-71 years) with histologically proven breast cancer between December 2009 and January 2011. PET-CT was performed at baseline and after NAC. SUV(max) percentage changes (ΔSUV(max)) were compared with pathology findings at surgery. Receiver-operator characteristic (ROC) analysis was used to discriminate between locoregional pCR and non-pCR. In patients not achieving pCR, it was investigated if ΔSUV(max) could accurately identify the residual cancer burden (RCB) classes: RCB-I (minimal residual disease (MRD)), RCB-II (moderate RD), and RCB-III (extensive RD). RESULTS pCR was obtained in 11 patients (25.6%). Residual disease was found in 32 patients (74.4%): 16 (37.2%) RCB-I, 15 (35.6%) RCB-II and 2 (4.7%) RCB-III. Sensitivity, specificity, and accuracy to predict pCR were 90.9%, 90.6%, and 90.7%, respectively. Specificity was 94.1% in the identification of a subset of patients who had either pCR or MRD. CONCLUSION Accuracy of ΔSUV(max) in the locoregional disease of stages II and III breast cancer patients after NAC is high for the identification of pCR cases. Its specificity is potentially sufficient to identify a subgroup of patients who could be managed with conservative surgery.
Collapse
Affiliation(s)
- Marta A García García-Esquinas
- Nuclear Medicine Department, Hospital Clínico San Carlos, Madrid, Spain; Radiology Department, Hospital Clínico San Carlos, Madrid, Spain.
| | | | | | - V Furió-Bacete
- Pathology Department, Hospital Clínico San Carlos, Madrid, Spain
| | | | | | | | | |
Collapse
|
46
|
Smith TAD, Zanda M, Fleming IN. Hypoxia stimulates 18F-fluorodeoxyglucose uptake in breast cancer cells via hypoxia inducible factor-1 and AMP-activated protein kinase. Nucl Med Biol 2013; 40:858-64. [PMID: 23786679 DOI: 10.1016/j.nucmedbio.2013.05.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 05/10/2013] [Accepted: 05/12/2013] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Hypoxia can stimulate (18)F-fluorodeoxyglucose (FDG) uptake in cultured cells. A better understanding of the underlying molecular mechanism is required to determine the value of FDG for studying tumour hypoxia. METHODS The effect of hypoxia on FDG uptake, and key proteins involved in glucose transport and glycolysis, was studied in MCF7 and MDA231 breast cancer cell lines. RESULTS Hypoxia induced a dose- and time-dependent increase in FDG uptake. The FDG increase was transient, suggesting that FDG uptake is only likely to be increased by acute hypoxia (<24 h). Molecular analysis indicated that hypoxia upregulated glut1 and 6-phosphofructo-2-kinase, key proteins involved in regulating glucose transport and glycolysis, and that these changes were induced by Hypoxia-Inducible factor 1 (HIF1) upregulation and/or AMP-activated protein kinase activation. CONCLUSIONS FDG may provide useful information about the oxygenation status of cells in hypoxic regions where HIF1 upregulation is hypoxia-driven.
Collapse
Affiliation(s)
- Tim A D Smith
- Division of Applied Medicine, School of Medicine and Dentistry, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD
| | | | | |
Collapse
|
47
|
Mghanga FP, Lan X, Bakari KH, Li C, Zhang Y. Fluorine-18 fluorodeoxyglucose positron emission tomography-computed tomography in monitoring the response of breast cancer to neoadjuvant chemotherapy: a meta-analysis. Clin Breast Cancer 2013; 13:271-9. [PMID: 23714689 DOI: 10.1016/j.clbc.2013.02.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 01/06/2013] [Accepted: 02/04/2013] [Indexed: 10/26/2022]
Abstract
INTRODUCTION To evaluate the diagnostic performance of fluorine-18 fluorodeoxyglucose positron emission tomography (FDG-PET) in monitoring the response of breast cancers to neoadjuvant chemotherapy. METHODS Articles published in medical and oncologic journals between January 2000 and June 2012 were identified by systematic MEDLINE, Cochrane Database for Systematic Reviews, and EMBASE, and by manual searches of the references listed in original and review articles. Quality of the included studies was assessed by using the quality assessment of diagnosis accuracy studies score tool. Meta-DiSc statistical software was used to calculate the summary sensitivity and specificity, positive predictive and negative predictive values, and the summary receiver operating characteristics curve (SROC). RESULTS Fifteen studies with 745 patients were included in the study after meeting the inclusion criteria. The pooled sensitivity and specificity of FDG-PET or PET/CT were 80.5% (95% CI, 75.9%-84.5%) and 78.8% (95% CI, 74.1%-83.0%), respectively, and the positive predictive and negative predictive values were 79.8% and 79.5%, respectively. After 1 and 2 courses of chemotherapy, the pooled sensitivity and false-positive rate were 78.2% (95% CI, 73.8%-82.5%) and 11.2%, respectively; and 82.4% (95% CI, 77.4%-86.1%) and 19.3%, respectively. CONCLUSIONS Analysis of the findings suggests that FDG-PET has moderately high sensitivity and specificity in early detection of responders from nonresponders, and can be applied in the evaluation of breast cancer response to neoadjuvant chemotherapy in patients with breast cancer.
Collapse
Affiliation(s)
- Fabian Pius Mghanga
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | | | | | | | | |
Collapse
|
48
|
Introduction to the analysis of PET data in oncology. J Pharmacokinet Pharmacodyn 2013; 40:419-36. [PMID: 23443280 DOI: 10.1007/s10928-013-9307-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 02/13/2013] [Indexed: 12/22/2022]
Abstract
Several reviews on specific topics related to positron emission tomography (PET) ranging in complexity from introductory to highly technical have already been published. This introduction to the analysis of PET data was written as a simple guide of the different phases of analysis of a given PET dataset, from acquisition to preprocessing, to the final data analysis. Although sometimes issues specific to PET in neuroimaging will be mentioned for comparison, most of the examples and applications provided will refer to oncology. Due to the limitations of space we couldn't address each issue comprehensively but, rather, we provided a general overview of each topic together with the references that the interested reader should consult. We will assume a familiarity with the basic principles of PET imaging.
Collapse
|
49
|
Is [18F] fluorodeoxyglucose uptake by the primary tumor a prognostic factor in breast cancer? Breast 2013; 22:39-43. [DOI: 10.1016/j.breast.2012.05.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 05/10/2012] [Accepted: 05/17/2012] [Indexed: 11/24/2022] Open
|
50
|
Marshall RP, Simpson JK, Lukey PT. Strategies for biomarker discovery in fibrotic disease. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1079-87. [PMID: 23376113 DOI: 10.1016/j.bbadis.2013.01.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Revised: 01/18/2013] [Accepted: 01/22/2013] [Indexed: 01/06/2023]
Abstract
The discovery and development of biomarkers for fibrotic diseases have potential utility in clinical decision-making as well as in pharmaceutical research and development. This review describes strategies for identifying diagnostic, prognostic and theranostic biomarkers. A range of technologies and platforms for biomarker discovery are highlighted, including several with specific relevance for fibrosis. Some challenges specific to fibrotic diseases are outlined including; benchmarking biomarkers against imperfect clinical measures of fibrosis, the complexity resulting from diverse aetiologies and target organs, and the availability of samples (including biopsy) from well-characterised patients with fibrotic disease. To overcome these challenges collaboration amongst clinical specialities as well as between academia and industry is essential. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.
Collapse
Affiliation(s)
- Richard P Marshall
- Fibrosis Discovery Performance Unit, GlaxoSmithKline R&D, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage SG1 2NY, UK.
| | | | | |
Collapse
|