1
|
Mouhanna P, Ståhlberg A, Andersson D, Albu-Kareem A, Elinder E, Eriksson O, Kavanagh A, Kovács A, Larsson KF, Linderholm B, Uminska M, Österlund T, Howell SJ, Ekholm M. Integration of personalised ultrasensitive ctDNA monitoring of patients with metastatic breast cancer to reduce imaging requirements. Int J Cancer 2024. [PMID: 39692755 DOI: 10.1002/ijc.35292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/21/2024] [Accepted: 11/28/2024] [Indexed: 12/19/2024]
Abstract
Circulating tumour DNA (ctDNA) is an emerging biomarker for monitoring cancers. The personalised disease monitoring in metastatic breast cancer (PDM-MBC) study is an ongoing study instigated to evaluate ctDNA as a biomarker to individualise imaging requirements in patients with MBC. Patients receiving first-line endocrine therapy (aromatase inhibitor + cyclin-dependent kinase 4/6 inhibitor) had plasma samples collected pre-treatment, weeks 2 and 4, and concurrently with imaging until progressive disease (PD). Here, we apply an experimental analytical workflow for ultrasensitive ctDNA analysis, utilising personalised ctDNA panels designed from mutations identified in tumour tissue, and present results for 24 patients. Twenty patients (83%) had detectable ctDNA pre-treatment. The median progression-free survival was 25.6 months, and 13 patients experienced PD, with rising ctDNA detected at or prior to PD in 12 patients (92%). If imaging had been omitted until the detection of rising ctDNA for at least one mutation, 68% (n = 71) of the scans performed amongst ctDNA-positive patients would have been avoided. Our results demonstrate that integration of personalised ctDNA monitoring of patients with MBC has potential to substantially reduce the imaging needs in patients showing ctDNA response to treatment.
Collapse
Affiliation(s)
- Pia Mouhanna
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Ryhov County Hospital, Jönköping, Sweden
| | - Anders Ståhlberg
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Daniel Andersson
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ahmed Albu-Kareem
- Department of Oncology, Linköping University Hospital, Linköping, Sweden
| | | | - Olle Eriksson
- Futurum - The Academy for Health and Care, Jönköping County, Sweden
| | - Amy Kavanagh
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Anikó Kovács
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Karolina F Larsson
- Sahlgrenska Center for Cancer Research, Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Barbro Linderholm
- Sahlgrenska Center for Cancer Research, Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Monika Uminska
- Department of Oncology, Kalmar County Hospital, Kalmar, Sweden
| | - Tobias Österlund
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Sacha J Howell
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Maria Ekholm
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Ryhov County Hospital, Jönköping, Sweden
- Department of Biomedical and Clinical Sciences, Division of Oncology, Linköping University, Linköping, Sweden
| |
Collapse
|
2
|
Merloni F, Palleschi M, Gianni C, Sirico M, Serra R, Casadei C, Sarti S, Cecconetto L, Di Menna G, Mariotti M, Maltoni R, Montanari D, Romeo A, De Giorgi U. Local treatment for oligoprogressive metastatic sites of breast cancer: efficacy, toxicities and future perspectives. Clin Exp Metastasis 2024; 41:863-875. [PMID: 39312051 PMCID: PMC11606987 DOI: 10.1007/s10585-024-10312-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/08/2024] [Indexed: 11/05/2024]
Abstract
Metastatic breast cancer (MBC) is still an incurable disease, which eventually develops resistance mechanisms against systemic therapies. While most patients experience widespread disease progression during systemic treatment (ST), in some cases, progression may occur at a limited number of metastatic sites. Evidence from other malignancies suggests that local treatment with stereotactic ablative radiotherapy (SABR) of oligoprogressive disease (OPD) may allow effective disease control without the need to modify ST. Available evidence regarding local treatment of oligoprogressive breast cancer is limited, mostly consisting of retrospective studies. The only randomized data come from the randomized CURB trial, which enrolled patients with oligoprogressive disease, including both small cell lung cancer and breast cancer patients, and did not show a survival benefit from local treatment in the latter group. However, local treatment of oligoprogressive MBC is still considered in clinical practice, especially to delay the switch to more toxic STs. This review aims to identify patients who may benefit from this approach based on the current available knowledge, focusing also on the potential risks associated with the combination of radiotherapy (RT) and ST, as well as on possible future scenarios.
Collapse
Affiliation(s)
- Filippo Merloni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy.
| | - Michela Palleschi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Caterina Gianni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Marianna Sirico
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Riccardo Serra
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Chiara Casadei
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Samanta Sarti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Lorenzo Cecconetto
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Giandomenico Di Menna
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Marita Mariotti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Roberta Maltoni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Daniela Montanari
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Antonino Romeo
- Radiotherapy Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| |
Collapse
|
3
|
Mu X, Li J, Huang J, Wang Z, Li Z, Li X, Jiang Y, Zhou Z, Fu W. 18F-NaF uptake in skull-base bone as a predictor of treatment response in advanced nasopharyngeal carcinoma. Sci Rep 2024; 14:29501. [PMID: 39604456 PMCID: PMC11603326 DOI: 10.1038/s41598-024-81350-w] [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: 09/23/2024] [Accepted: 11/26/2024] [Indexed: 11/29/2024] Open
Abstract
This study investigates the utility of 18F-sodium fluoride (18F-NaF) positron emission tomography/computed tomography (PET/CT) in assessing skull-base bone invasion (SBBI) and predicting treatment response in advanced nasopharyngeal carcinoma (NPC). A retrospective analysis was conducted on 142 patients with newly diagnosed advanced NPC who underwent 18F-NaF PET/CT for initial staging from December 2020 to December 2023. 18F-NaF PET/CT scans were analyzed for uptake values at the skull-base bone, and these were correlated with treatment outcomes of primary tumor using the Response Evaluation Criteria in Solid Tumors (RECIST) 1.1. Statistical analyses involved Mann-Whitney U tests for group comparisons and logistic regression for evaluating risk factors. Higher 18F-NaF uptake at the skull-base bone was significantly associated with advanced T stages (p < 0.0001) and the presence of bone metastases (p = 0.01). Patients exhibiting complete response (CR) to treatment had significantly lower skull-base 18F-NaF uptake compared to those with non-CR (p < 0.001). Receiver operating characteristic (ROC) analysis identified an SUVmax > 10.0 and SUVmean > 5.2 as predictive of non-CR, with AUC values of 0.77 and 0.76, respectively. Univariate and multivariable analysis confirmed SUVmax as a significant predictor of treatment response (OR = 7.03, 95% CI: 1.97-25.13, p < 0.05). Elevated 18F-NaF uptake at the skull-base bone is predictive of poorer treatment outcomes, highlighting its potential as a prognostic biomarker in advanced NPC. This study demonstrates that 18F-NaF PET/CT is a valuable imaging modality for evaluating SBBI in NPC, offering metabolic information that complements the anatomical findings from MRI.
Collapse
Affiliation(s)
- Xingyu Mu
- Department of Nuclear Medicine, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Jingze Li
- Department of Nuclear Medicine, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Jingquan Huang
- Department of Nuclear Medicine, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Zhenzhen Wang
- Department of Nuclear Medicine, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Zuguo Li
- Department of Nuclear Medicine, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xun Li
- Department of Radiology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Yu Jiang
- Department of Radiology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Zhipeng Zhou
- Department of Radiology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi Zhuang Autonomous Region, People's Republic of China.
| | - Wei Fu
- Department of Nuclear Medicine, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi Zhuang Autonomous Region, People's Republic of China.
| |
Collapse
|
4
|
Vaz SC, Woll JPP, Cardoso F, Groheux D, Cook GJR, Ulaner GA, Jacene H, Rubio IT, Schoones JW, Peeters MJV, Poortmans P, Mann RM, Graff SL, Dibble EH, de Geus-Oei LF. Joint EANM-SNMMI guideline on the role of 2-[ 18F]FDG PET/CT in no special type breast cancer : (endorsed by the ACR, ESSO, ESTRO, EUSOBI/ESR, and EUSOMA). Eur J Nucl Med Mol Imaging 2024; 51:2706-2732. [PMID: 38740576 PMCID: PMC11224102 DOI: 10.1007/s00259-024-06696-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/20/2024] [Indexed: 05/16/2024]
Abstract
INTRODUCTION There is much literature about the role of 2-[18F]FDG PET/CT in patients with breast cancer (BC). However, there exists no international guideline with involvement of the nuclear medicine societies about this subject. PURPOSE To provide an organized, international, state-of-the-art, and multidisciplinary guideline, led by experts of two nuclear medicine societies (EANM and SNMMI) and representation of important societies in the field of BC (ACR, ESSO, ESTRO, EUSOBI/ESR, and EUSOMA). METHODS Literature review and expert discussion were performed with the aim of collecting updated information regarding the role of 2-[18F]FDG PET/CT in patients with no special type (NST) BC and summarizing its indications according to scientific evidence. Recommendations were scored according to the National Institute for Health and Care Excellence (NICE) criteria. RESULTS Quantitative PET features (SUV, MTV, TLG) are valuable prognostic parameters. In baseline staging, 2-[18F]FDG PET/CT plays a role from stage IIB through stage IV. When assessing response to therapy, 2-[18F]FDG PET/CT should be performed on certified scanners, and reported either according to PERCIST, EORTC PET, or EANM immunotherapy response criteria, as appropriate. 2-[18F]FDG PET/CT may be useful to assess early metabolic response, particularly in non-metastatic triple-negative and HER2+ tumours. 2-[18F]FDG PET/CT is useful to detect the site and extent of recurrence when conventional imaging methods are equivocal and when there is clinical and/or laboratorial suspicion of relapse. Recent developments are promising. CONCLUSION 2-[18F]FDG PET/CT is extremely useful in BC management, as supported by extensive evidence of its utility compared to other imaging modalities in several clinical scenarios.
Collapse
Affiliation(s)
- Sofia C Vaz
- Nuclear Medicine-Radiopharmacology, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal.
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.
| | | | - Fatima Cardoso
- Breast Unit, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
| | - David Groheux
- Nuclear Medicine Department, Saint-Louis Hospital, Paris, France
- University Paris-Diderot, INSERM U976, Paris, France
- Centre d'Imagerie Radio-Isotopique (CIRI), La Rochelle, France
| | - Gary J R Cook
- Department of Cancer Imaging, King's College London, London, UK
- King's College London and Guy's & St Thomas' PET Centre, London, UK
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Gary A Ulaner
- Molecular Imaging and Therapy, Hoag Family Cancer Institute, Newport Beach, CA, USA
- University of Southern California, Los Angeles, CA, USA
| | - Heather Jacene
- Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Isabel T Rubio
- Breast Surgical Oncology, Clinica Universidad de Navarra, Madrid, Cancer Center Clinica Universidad de Navarra, Navarra, Spain
| | - Jan W Schoones
- Directorate of Research Policy, Leiden University Medical Center, Leiden, The Netherlands
| | - Marie-Jeanne Vrancken Peeters
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Philip Poortmans
- Department of Radiation Oncology, Iridium Netwerk, Antwerp, Belgium
- University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Ritse M Mann
- Radiology Department, RadboudUMC, Nijmegen, The Netherlands
| | - Stephanie L Graff
- Lifespan Cancer Institute, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Elizabeth H Dibble
- Department of Diagnostic Imaging, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA.
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.
- Biomedical Photonic Imaging Group, University of Twente, Enschede, The Netherlands.
- Department of Radiation Science & Technology, Technical University of Delft, Delft, The Netherlands.
| |
Collapse
|
5
|
Teng X, Han K, Jin W, Ma L, Wei L, Min D, Chen L, Du Y. Development and validation of an early diagnosis model for bone metastasis in non-small cell lung cancer based on serological characteristics of the bone metastasis mechanism. EClinicalMedicine 2024; 72:102617. [PMID: 38707910 PMCID: PMC11066529 DOI: 10.1016/j.eclinm.2024.102617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 05/07/2024] Open
Abstract
Background Bone metastasis significantly impact the prognosis of non-small cell lung cancer (NSCLC) patients, reducing their quality of life and shortening their survival. Currently, there are no effective tools for the diagnosis and risk assessment of early bone metastasis in NSCLC patients. This study employed machine learning to analyze serum indicators that are closely associated with bone metastasis, aiming to construct a model for the timely detection and prognostic evaluation of bone metastasis in NSCLC patients. Methods The derivation cohort consisted of 664 individuals with stage IV NSCLC, diagnosed between 2015 and 2018. The variables considered in this study included age, sex, and 18 specific serum indicators that have been linked to the occurrence of bone metastasis in NSCLC. Variable selection used multivariate logistic regression analysis and Lasso regression analysis. Six machine learning methods were utilized to develop a bone metastasis diagnostic model, assessed with Area Under the Curve (AUC), Decision Curve Analysis (DCA), sensitivity, specificity, and validation cohorts. External validation used 113 NSCLC patients from the Medical Alliance (2019-2020). Furthermore, a prospective validation study was conducted on a cohort of 316 patients (2019-2020) who were devoid of bone metastasis, and followed-up for at least two years to assess the predictive capabilities of this model. The model's prognostic value was evaluated using Kaplan-Meier survival curves. Findings Through variable selection, 11 serum indictors were identified as independent predictive factors for NSCLC bone metastasis. Six machine learning models were developed using age, sex, and these serum indicators. A random forest (RF) model demonstrated strong performance during the training and internal validation cohorts, achieving an AUC of 0.98 (95% CI 0.95-0.99) for internal validation. External validation further confirmed the RF model's effectiveness, yielding an AUC of 0.97 (95% CI 0.94-0.99). The calibration curves demonstrated a high level of concordance between the anticipated risk and the observed risk of the RF model. Prospective validation revealed that the RF model could predict the occurrence of bone metastasis approximately 10.27 ± 3.58 months in advance, according to the results of the SPECT. An online computing platform (https://bonemetastasis.shinyapps.io/shiny_cls_1model/) for this RF model is publicly available and free-to-use by doctors and patients. Interpretation This study innovatively employs age, gender, and 11 serological markers closely related to the mechanism of bone metastasis to construct an RF model, providing a reliable tool for the early screening and prognostic assessment of bone metastasis in NSCLC patients. However, as an exploratory study, the findings require further validation through large-scale, multicenter prospective studies. Funding This work is supported by the National Natural Science Foundation of China (NO.81974315); Shanghai Municipal Science and Technology Commission Medical Innovation Research Project (NO.20Y11903300); Shanghai Municipal Health Commission Health Industry Clinical Research Youth Program (NO.20204Y034).
Collapse
Affiliation(s)
- Xiaoyan Teng
- Department of Laboratory Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Kun Han
- Department of Oncology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Wei Jin
- Department of Laboratory Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Liru Ma
- Department of Laboratory Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Lirong Wei
- Department of Laboratory Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Daliu Min
- Department of Oncology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Libo Chen
- Department of Nuclear Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yuzhen Du
- Department of Laboratory Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| |
Collapse
|
6
|
Zamani-Siahkali N, Mirshahvalad SA, Farbod A, Divband G, Pirich C, Veit-Haibach P, Cook G, Beheshti M. SPECT/CT, PET/CT, and PET/MRI for Response Assessment of Bone Metastases. Semin Nucl Med 2024; 54:356-370. [PMID: 38172001 DOI: 10.1053/j.semnuclmed.2023.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 01/05/2024]
Abstract
Recent developments in hybrid SPECT/CT systems and the use of cadmium-zinc-telluride (CZT) detectors have improved the diagnostic accuracy of bone scintigraphy. These advancements have paved the way for novel quantitative approaches to accurate and reproducible treatment monitoring of bone metastases. PET/CT imaging using [18F]F-FDG and [18F]F-NaF have shown promising clinical utility in bone metastases assessment and monitoring response to therapy and prediction of treatment response in a broad range of malignancies. Additionally, specific tumor-targeting tracers like [99mTc]Tc-PSMA, [68Ga]Ga-PSMA, or [11C]C- or [18F]F-Choline revealed high diagnostic performance for early assessment and prognostication of bone metastases, particularly in prostate cancer. PET/MRI appears highly accurate imaging modality, but has associated limitations notably, limited availability, more complex logistics and high installation costs. Advances in artificial intelligence (Al) seem to improve the accuracy of imaging modalities and provide an assistant role in the evaluation of treatment response of bone metastases.
Collapse
Affiliation(s)
- Nazanin Zamani-Siahkali
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria; Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Mirshahvalad
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria; Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, Toronto, Canada
| | - Abolfazl Farbod
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria; Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Christian Pirich
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria
| | - Patrick Veit-Haibach
- Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, Toronto, Canada
| | - Gary Cook
- Cancer Imaging Department, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Mohsen Beheshti
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
7
|
Ulaner GA, Vaz SC, Groheux D. Quarter-Century Transformation of Oncology: Positron Emission Tomography for Patients with Breast Cancer. PET Clin 2024; 19:147-162. [PMID: 38177052 DOI: 10.1016/j.cpet.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
PET radiotracers have become indispensable in the care of patients with breast cancer. 18F-fluorodeoxyglucose has become the preferred method of many oncologists for systemic staging of breast cancer at initial diagnosis, detecting recurrent disease, and for measuring treatment response after therapy. 18F-Sodium Fluoride is valuable for detection of osseous metastases. 18F-fluoroestradiol is now FDA-approved with multiple appropriate clinical uses. There are multiple PET radiotracers in clinical trials, which may add utility of PET imaging for patients with breast cancer in the future. This article will describe the advances during the last quarter century in PET for patients with breast cancer.
Collapse
Affiliation(s)
- Gary A Ulaner
- Molecular Imaging and Therapy, Hoag Family Cancer Institute, Irvine, CA, USA; Departments of Radiology and Translational Genomics, University of Southern California, Los Angeles, CA, USA.
| | - Sofia Carrilho Vaz
- Nuclear Medicine-Radiopharmacology, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - David Groheux
- Nuclear Department of Nuclear Medicine, Saint-Louis Hospital, Paris, France; Centre d'Imagerie Radio-Isotopique (CIRI), La Rochelle, France; University Paris-Diderot, INSERM U976, HIPI, Paris, France
| |
Collapse
|
8
|
Muzi M, Peterson LM, Specht JM, Hippe DS, Novakova-Jiresova A, Lee JH, Kurland BF, Mankoff DA, Obuchowski N, Linden HM, Kinahan PE. Repeatability of 18F-FDG uptake in metastatic bone lesions of breast cancer patients and implications for accrual to clinical trials. EJNMMI Res 2024; 14:32. [PMID: 38536511 PMCID: PMC10973316 DOI: 10.1186/s13550-024-01093-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/06/2024] [Indexed: 04/01/2024] Open
Abstract
BACKGROUND Standard measures of response such as Response Evaluation Criteria in Solid Tumors are ineffective for bone lesions, often making breast cancer patients that have bone-dominant metastases ineligible for clinical trials with potentially helpful therapies. In this study we prospectively evaluated the test-retest uptake variability of 2-deoxy-2-[18F]fluoro-D-glucose (18F-FDG) in a cohort of breast cancer patients with bone-dominant metastases to determine response criteria. The thresholds for 95% specificity of change versus no-change were then applied to a second cohort of breast cancer patients with bone-dominant metastases. METHODS For this study, nine patients with 38 bone lesions were imaged with 18F-FDG in the same calibrated scanner twice within 14 days. Tumor uptake was quantified by the most commonly used PET parameter, the maximum tumor voxel normalized by dose and body weight (SUVmax) and also by the mean of a 1-cc maximal uptake volume normalized by dose and lean-body-mass (SULpeak). The asymmetric repeatability coefficients with confidence intervals for SUVmax and SULpeak were used to determine the limits of 18F-FDG uptake variability. A second cohort of 28 breast cancer patients with bone-dominant metastases that had 146 metastatic bone lesions was imaged with 18F-FDG before and after standard-of-care therapy for response assessment. RESULTS The mean relative difference of SUVmax and SULpeak in 38 bone tumors of the first cohort were 4.3% and 6.7%. The upper and lower asymmetric limits of the repeatability coefficient were 19.4% and - 16.3% for SUVmax, and 21.2% and - 17.5% for SULpeak. 18F-FDG repeatability coefficient confidence intervals resulted in the following patient stratification using SULpeak for the second patient cohort: 11-progressive disease, 5-stable disease, 7-partial response, and 1-complete response with three inevaluable patients. The asymmetric repeatability coefficients response criteria for SULpeak changed the status of 3 patients compared to the standard Positron Emission Tomography Response Criteria in Solid Tumors of ± 30% SULpeak. CONCLUSION In evaluating bone tumor response for breast cancer patients with bone-dominant metastases using 18F-FDG SUVmax, the repeatability coefficients from test-retest studies show that reductions of more than 17% and increases of more than 20% are unlikely to be due to measurement variability. Serial 18F-FDG imaging in clinical trials investigating bone lesions in these patients, such as the ECOG-ACRIN EA1183 trial, benefit from confidence limits that allow interpretation of response.
Collapse
Affiliation(s)
- Mark Muzi
- Department of Radiology, University of Washington Medical Center, 1959 NE Pacific Street, UW Box 356465, Seattle, Washington, 98195, USA.
| | - Lanell M Peterson
- Department of Radiology, University of Washington Medical Center, 1959 NE Pacific Street, UW Box 356465, Seattle, Washington, 98195, USA
| | - Jennifer M Specht
- Department of Radiology, University of Washington Medical Center, 1959 NE Pacific Street, UW Box 356465, Seattle, Washington, 98195, USA
| | - Daniel S Hippe
- Department of Radiology, University of Washington Medical Center, 1959 NE Pacific Street, UW Box 356465, Seattle, Washington, 98195, USA
| | | | - Jean H Lee
- Department of Radiology, University of Washington Medical Center, 1959 NE Pacific Street, UW Box 356465, Seattle, Washington, 98195, USA
| | - Brenda F Kurland
- Department of Radiology, University of Washington Medical Center, 1959 NE Pacific Street, UW Box 356465, Seattle, Washington, 98195, USA
| | | | | | - Hannah M Linden
- Department of Radiology, University of Washington Medical Center, 1959 NE Pacific Street, UW Box 356465, Seattle, Washington, 98195, USA
| | - Paul E Kinahan
- Department of Radiology, University of Washington Medical Center, 1959 NE Pacific Street, UW Box 356465, Seattle, Washington, 98195, USA
| |
Collapse
|
9
|
Jacene H, Dietsche E, Specht J. The Current and Future Roles of Precision Oncology in Advanced Breast Cancer. J Nucl Med 2024; 65:349-356. [PMID: 38302151 DOI: 10.2967/jnumed.122.264882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 02/03/2024] Open
Abstract
Breast cancer is a common but heterogeneous disease characterized by several biologic features, including tumor grade, hormone receptor status, human epidermal growth factor receptor 2 status, and gene expression assays. These biologic and genomic features drive treatment decisions. In the advanced disease setting, inter- and intrapatient tumor heterogeneity is increasingly recognized as a challenge for optimizing treatment. Recent evidence and the recent approval of novel radiopharmaceuticals have increased recognition and acceptance of the potential of molecular imaging as a biomarker to impact and guide management decisions for advanced breast cancer.
Collapse
Affiliation(s)
- Heather Jacene
- Imaging/Radiology, Dana-Farber/Brigham Cancer Center, Boston, Massachusetts;
| | - Eric Dietsche
- Department of Radiology, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island; and
| | - Jennifer Specht
- Fred Hutch Cancer Center, Divisions of Hematology and Oncology and of Clinical Research, Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
10
|
Muzi M, Peterson LM, Specht JM, Hippe DS, Novakova-Jiresova A, Lee JH, Kurland BF, Mankoff DA, Obuchowski N, Linden HM, Kinahan PE. Repeatability of 18F-FDG uptake in metastatic bone lesions of breast cancer patients and implications for accrual to clinical trials. RESEARCH SQUARE 2024:rs.3.rs-3818932. [PMID: 38313279 PMCID: PMC10836099 DOI: 10.21203/rs.3.rs-3818932/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
BACKGROUND Standard measures of response such as Response Evaluation Criteria in Solid Tumors are ineffective for bone lesions, often making breast cancer patients with bone-dominant metastases ineligible for clinical trials with potentially helpful therapies. In this study we prospectively evaluated the test-retest uptake variability of 2-deoxy-2-[18F]fluoro-D-glucose (18F-FDG) in a cohort of breast cancer patients with bone-dominant metastases to determine response criteria. The thresholds for 95% specificity of change versus no-change were then applied to a second cohort of breast cancer patients with bone-dominant metastases.In this study, nine patients with 38 bone lesions were imaged with 18F-FDG in the same calibrated scanner twice within 14 days. Tumor uptake was quantified as the maximum tumor voxel normalized by dose and body weight (SUVmax) and the mean of a 1-cc maximal uptake volume normalized by dose and lean-body-mass (SULpeak). The asymmetric repeatability coefficients with confidence intervals of SUVmax and SULpeak were used to determine limits of 18F-FDG uptake variability. A second cohort of 28 breast cancer patients with bone-dominant metastases that had 146 metastatic bone lesions was imaged with 18F-FDG before and after standard-of-care therapy for response assessment. RESULTS The mean relative difference of SUVmax in 38 bone tumors of the first cohort was 4.3%. The upper and lower asymmetric limits of the repeatability coefficient were 19.4% and -16.3%, respectively. The 18F-FDG repeatability coefficient confidence intervals resulted in the following patient stratification for the second patient cohort: 11-progressive disease, 5-stable disease, 7-partial response, and 1-complete response with three inevaluable patients. The asymmetric repeatability coefficients response criteria changed the status of 3 patients compared to standard the standard Positron Emission Tomography Response Criteria in Solid Tumors of ±30% SULpeak. CONCLUSIONS In evaluating bone tumor response for breast cancer patients with bone-dominant metastases using 18F-FDG uptake, the repeatability coefficients from test-retest studies show that reductions of more than 17% and increases of more than 20% are unlikely to be due to measurement variability. Serial 18F-FDG imaging in clinical trials investigating bone lesions from these patients, such as the ECOG-ACRIN EA1183 trial, benefit from confidence limits that allow interpretation of response.
Collapse
Affiliation(s)
- Mark Muzi
- University of Washington School of Medicine
| | | | | | | | | | - Jean H Lee
- University of Washington Department of Radiology
| | | | | | | | | | | |
Collapse
|
11
|
Ouvrard E, Kaseb A, Poterszman N, Porot C, Somme F, Imperiale A. Nuclear medicine imaging for bone metastases assessment: what else besides bone scintigraphy in the era of personalized medicine? Front Med (Lausanne) 2024; 10:1320574. [PMID: 38288299 PMCID: PMC10823373 DOI: 10.3389/fmed.2023.1320574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/28/2023] [Indexed: 01/31/2024] Open
Abstract
Accurate detection and reliable assessment of therapeutic responses in bone metastases are imperative for guiding treatment decisions, preserving quality of life, and ultimately enhancing overall survival. Nuclear imaging has historically played a pivotal role in this realm, offering a diverse range of radiotracers and imaging modalities. While the conventional bone scan using 99mTc marked bisphosphonates has remained widely utilized, its diagnostic performance is hindered by certain limitations. Positron emission tomography, particularly when coupled with computed tomography, provides improved spatial resolution and diagnostic performance with various pathology-specific radiotracers. This review aims to evaluate the performance of different nuclear imaging modalities in clinical practice for detecting and monitoring the therapeutic responses in bone metastases of diverse origins, addressing their limitations and implications for image interpretation.
Collapse
Affiliation(s)
- Eric Ouvrard
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Ashjan Kaseb
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
- Radiology, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Nathan Poterszman
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Clémence Porot
- Radiopharmacy, Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France
| | - Francois Somme
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Alessio Imperiale
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
- IPHC, UMR 7178, CNRS/Unistra, Strasbourg, France
| |
Collapse
|
12
|
Yağar H, Aytekin MN, Şener Dede D, Şendur MAN, Öztürk R, Yalçın B. Do metastatic volumes measured in breast cancer patients with bone metastases correlate with the numbers of skeletal and extraskeletal events? Jt Dis Relat Surg 2024; 35:105-111. [PMID: 38108171 PMCID: PMC10746892 DOI: 10.52312/jdrs.2023.1333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 10/02/2023] [Indexed: 12/19/2023] Open
Abstract
OBJECTIVES The study aimed to investigate the relationship between metastatic volume measurement, skeletal-related events, and survival in women diagnosed with breast cancer and bone metastases. PATIENTS AND METHODS This retrospective study was conducted with 82 female breast cancer patients (mean age: 53±14.3 years; range, 23 to 87 years) diagnosed, treated, and followed up between January 2005 and December 2019. The collected data included information on metastasis sites and the presence of skeletal-related events. Metastatic volume was measured in two ways: the number of metastases (high to low) and their localization (the first, second, and third groups). The first group consisted of vertebrae, ribs, sternum, and calvarial bones; the second group included scapula, clavicle, proximal humerus, and proximal femur regions; the third group consisted of femur and humerus diaphyseal and distal regions, as well as metastasis regions in other long bones. RESULTS Sixty-three (76.8%) patients were diagnosed with ductal carcinoma. Half of the patients had bone metastases at the time of initial diagnosis, while 62 (75.6%) experienced skeletal-related events, with at least three events occurring in 30 (36.6%) patients. Bone pain was the most common skeletal-related event. No correlation was found between metastatic volume measurement based on the localization of bone metastases and the number of bones and the occurrence of skeletal-related events (p>0.05 for each). Patients' survival time spanned from one to 231 months (median: 56.8 months) from their first diagnosis. Patients with high metastatic volume, those in the third group, those whose pelvis and lung were involved, and elderly patients had a shorter survival time (p<0.05 for each). CONCLUSION The study indicates that measuring metastatic volume may be a critical factor in evaluating the survival of breast cancer patients with bone metastases. Future prospective and randomized controlled studies can explore the potential of this measurement to create practical clinical tools.
Collapse
Affiliation(s)
| | | | | | | | - Recep Öztürk
- Dr. Abdurrahman Yurtaslan Ankara Onkoloji Eğitim Araştırma Hastanesi, Ortopedi ve Travmatoloji Kliniği, 06200 Yenimahalle, Ankara, Türkiye.
| | | |
Collapse
|
13
|
Castorina L, Comis AD, Prestifilippo A, Quartuccio N, Panareo S, Filippi L, Castorina S, Giuffrida D. Innovations in Positron Emission Tomography and State of the Art in the Evaluation of Breast Cancer Treatment Response. J Clin Med 2023; 13:154. [PMID: 38202160 PMCID: PMC10779934 DOI: 10.3390/jcm13010154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/14/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
The advent of hybrid Positron Emission Tomography/Computed Tomography (PET/CT) and PET/Magnetic Resonance Imaging (MRI) scanners resulted in an increased clinical relevance of nuclear medicine in oncology. The use of [18F]-Fluorodeoxyglucose ([18F]FDG) has also made it possible to study tumors (including breast cancer) from not only a dimensional perspective but also from a metabolic point of view. In particular, the use of [18F]FDG PET allowed early confirmation of the efficacy or failure of therapy. The purpose of this review was to assess the literature concerning the response to various therapies for different subtypes of breast cancer through PET. We start by summarizing studies that investigate the validation of PET/CT for the assessment of the response to therapy in breast cancer; then, we present studies that compare PET imaging (including PET devices dedicated to the breast) with CT and MRI, focusing on the identification of the most useful parameters obtainable from PET/CT. We also focus on novel non-FDG radiotracers, as they allow for the acquisition of information on specific aspects of the new therapies.
Collapse
Affiliation(s)
- Luigi Castorina
- Nuclear Medicine Outpatient Unit, REM Radiotherapy Srl, Via Penninanzzo 11, 95029 Viagrande, Italy;
| | - Alessio Danilo Comis
- Nuclear Medicine Outpatient Unit, REM Radiotherapy Srl, Via Penninanzzo 11, 95029 Viagrande, Italy;
| | - Angela Prestifilippo
- Department of Oncology, IOM Mediterranean Oncology Institute, Via Penninanzzo 7, 95029 Viagrande, Italy; (A.P.); (D.G.)
| | - Natale Quartuccio
- Nuclear Medicine Unit, Ospedali Riuniti Villa Sofia-Cervello, 90146 Palermo, Italy;
| | - Stefano Panareo
- Nuclear Medicine Unit, Oncology and Haematology Department, University Hospital of Modena, 41124 Modena, Italy;
| | - Luca Filippi
- Nuclear Medicine Unit, Department of Oncohaematology, Fondazione PTV Policlinico Tor Vergata University Hospital, Viale Oxford 81, 00133 Rome, Italy;
| | - Serena Castorina
- Nuclear Medicine Unit, Azienda Ospedaliero Universitaria Policlinico “G. Rodolico-San Marco”, 95123 Catania, Italy
| | - Dario Giuffrida
- Department of Oncology, IOM Mediterranean Oncology Institute, Via Penninanzzo 7, 95029 Viagrande, Italy; (A.P.); (D.G.)
| |
Collapse
|
14
|
Ozawa A, Iwasaki M, Yokoyama K, Tsuchiya J, Kawano R, Nishihara H, Tateishi U. Correlation between choline kinase alpha expression and 11C-choline accumulation in breast cancer using positron emission tomography/computed tomography: a retrospective study. Sci Rep 2023; 13:17620. [PMID: 37848481 PMCID: PMC10582087 DOI: 10.1038/s41598-023-44542-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/10/2023] [Indexed: 10/19/2023] Open
Abstract
Choline kinase (CK) is reportedly overexpressed in various malignancies. Among its isoforms, CKα overexpression is presumably related to oncogenic change. Choline positron emission tomography (PET) is reportedly useful for detecting and evaluating therapy outcomes in malignancies. In this study, we investigated the correlation between CKα expression and 11C-choline accumulation in breast cancer cells. We also compared the CKα expression level with other pathological findings for investigating tumour activity. Fifty-six patients with breast cancer (mean age: 51 years) who underwent their first medical examination between May 2007 and December 2008 were enrolled. All the patients underwent 11C-choline PET/computed tomography imaging prior to surgery. The maximum standardised uptake value was recorded for evaluating 11C-choline accumulation. The intensity of CKα expression was classified using immunostaining. A significant correlation was observed between CKα expression and 11C-choline accumulation (P < 0.0001). A comparison of breast cancer mortality demonstrated that strong CKα expression was associated with a shorter survival time (P < 0.0001). 11C-choline accumulation was also negatively correlated with survival time (P < 0.0001). Tumours with strong CKα expression are reportedly highly active in breast cancer. A correlation was observed between CKα expression and 11C-choline accumulation, suggesting their role as prognostic indicators of breast cancer.
Collapse
Affiliation(s)
- Akane Ozawa
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Masako Iwasaki
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Kota Yokoyama
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Junichi Tsuchiya
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Ryutaro Kawano
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Nishihara
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Ukihide Tateishi
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan.
| |
Collapse
|
15
|
Hardcastle N, Liu Y, Siva S, David S. [ 18F]NaF PET/CT imaging of response to single fraction SABR to bone metastases from breast cancer. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2023; 3:1197397. [PMID: 39380960 PMCID: PMC11460292 DOI: 10.3389/fnume.2023.1197397] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 09/15/2023] [Indexed: 10/10/2024]
Abstract
Breast cancer commonly metastasises to the skeleton, and stereotactic ablative body radiation therapy (SABR) is an emerging treatment for oligometastatic disease. Accurately imaging bone metastases and their response to treatment is challenging. [18F]NaF-PET has a higher sensitivity and specificity than conventional bone scans for detecting breast cancer bone metastases. In this pre-defined secondary analysis of a prospective trial, we evaluated the change in [18F]NaF uptake after SABR. Patients with oligometastatic breast cancer received a single fraction of 20 Gy to up to three bone metastases. [18F]NaF-PET was acquired before and 12 months after SABR. Pre- and post-treatment [18F]NaF-PET images were registered to the treatment planning CT. The relative change in tumour SUVmax and SUVmean was quantified. The intersection of each of the radiation therapy isodose contours with a non-tumour bone was created. The change in SUVmean in sub-volumes of non-tumour bone receiving doses of 0-20 Gy was quantified. In total, 14 patients, with 17 bone metastases, were available for analysis. A total of 15 metastases exhibited a reduction in SUVmax; the median reduction was 42% and the maximum reduction 82%. An increased absolute reduction in SUVmax was observed with higher pre-treatment SUVmax. One patient exhibited increased SUVmax after treatment, which was attributed to normal peri-tumoural bone regeneration in the context of a bone metastasis. There was a median reduction of 15%-34% for non-tumour bone in each dose level.
Collapse
Affiliation(s)
- Nicholas Hardcastle
- Department of Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Centre for Medical Radiation Physics, University of Wollongong, Wollongong, NSW, Australia
| | - Yang Liu
- Western Health Victoria, Melbourne, VIC, Australia
| | - Shankar Siva
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Steven David
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| |
Collapse
|
16
|
Abstract
Breast cancer (BC) remains one of the leading causes of death among women. The management and outcome in BC are strongly influenced by a multidisciplinary approach, which includes available treatment options and different imaging modalities for accurate response assessment. Among breast imaging modalities, MR imaging is the modality of choice in evaluating response to neoadjuvant therapy, whereas F-18 Fluorodeoxyglucose positron emission tomography, conventional computed tomography (CT), and bone scan play a vital role in assessing response to therapy in metastatic BC. There is an unmet need for a standardized patient-centric approach to use different imaging methods for response assessment.
Collapse
Affiliation(s)
- Saima Muzahir
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Imaging Sciences, 1364 Clifton Road, Atlanta GA 30322, USA; Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Imaging Sciences, Emory University Hospital, Room E152, 1364 Clifton Road, Atlanta, GA 30322, USA.
| | - Gary A Ulaner
- Molecular Imaging and Therapy, Hoag Family Cancer Institute, Newport Beach, CA, USA; Radiology and Translational Genomics, University of Southern California, Los Angeles, CA, USA
| | - David M Schuster
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Imaging Sciences, Emory University Hospital, Room E152, 1364 Clifton Road, Atlanta, GA 30322, USA
| |
Collapse
|
17
|
Khojasteh E, Dehdashti F, Shokeen M. Molecular imaging of bone metastasis. J Bone Oncol 2023; 40:100477. [PMID: 37193117 PMCID: PMC10182320 DOI: 10.1016/j.jbo.2023.100477] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 05/18/2023] Open
Abstract
Recent advances in molecularly targeted modular designs for in vivo imaging applications has thrusted open possibilities of investigating deep molecular interactions non-invasively and dynamically. The shifting landscape of biomarker concentration and cellular interactions throughout pathological progression requires quick adaptation of imaging agents and detection modalities for accurate readouts. The synergy of state of art instrumentation with molecularly targeted molecules is resulting in more precise, accurate and reproducible data sets, which is facilitating investigation of several novel questions. Small molecules, peptides, antibodies and nanoparticles are some of the commonly used molecular targeting vectors that can be applied for imaging as well as therapy. The field of theranostics, which encompasses joint application of therapy and imaging, is successfully leveraging the multifunctional use of these biomolecules [[1], [2]]. Sensitive detection of cancerous lesions and accurate assessment of treatment response has been transformative for patient management. Particularly, since bone metastasis is one of the dominant causes of morbidity and mortality in cancer patients, imaging can be hugely impactful in this patient population. The intent of this review is to highlight the utility of molecular positron emission tomography (PET) imaging in the context of prostate and breast bone metastatic cancer, and multiple myeloma. Furthermore, comparisons are drawn with traditionally utilized bone scans (skeletal scintigraphy). Both these modalities can be synergistic or complementary for assessing lytic- and blastic- bone lesions.
Collapse
Affiliation(s)
- Eliana Khojasteh
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Farrokh Dehdashti
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Monica Shokeen
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
- Corresponding author at: Mallinckrodt Institute of Radiology, 510 South Kingshighway Boulevard, St. Louis, MO 63110, USA.
| |
Collapse
|
18
|
van Geel JJL, de Vries EFJ, van Kruchten M, Hospers GAP, Glaudemans AWJM, Schröder CP. Molecular imaging as biomarker for treatment response and outcome in breast cancer. Ther Adv Med Oncol 2023; 15:17588359231170738. [PMID: 37223262 PMCID: PMC10201167 DOI: 10.1177/17588359231170738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 03/28/2023] [Indexed: 05/25/2023] Open
Abstract
Molecular imaging, such as positron emission tomography (PET), is increasingly used as biomarker to predict and assess treatment response in breast cancer. The number of biomarkers is expanding with specific tracers for tumour characteristics throughout the body and this information can be used to aid the decision-making process. These measurements include metabolic activity using [18F]fluorodeoxyglucose PET ([18F]FDG-PET), oestrogen receptor (ER) expression using 16α-[18F]Fluoro-17β-oestradiol ([18F]FES)-PET and human epidermal growth factor receptor 2 (HER2) expression using PET with radiolabelled trastuzumab (HER2-PET). In early breast cancer, baseline [18F]FDG-PET is frequently used for staging, but limited subtype-specific data reduce its usefulness as biomarker for treatment response or outcome. Early metabolic change on serial [18F]FDG-PET is increasingly used in the neo-adjuvant setting as dynamic biomarker to predict pathological complete response to systemic therapy, potentially allowing de-intensification or step-up intensification of treatment. In the metastatic setting, baseline [18F]FDG-PET and [18F]FES-PET can be used as biomarker to predict treatment response, in triple-negative and ER-positive breast cancer, respectively. Metabolic progression on repeated [18F]FDG-PET appears to precede progressive disease on standard evaluation imaging; however, subtype-specific studies are limited and more prospective data are needed before implementation in clinical practice. Even though (repeated) [18F]FDG-PET, [18F]FES-PET and HER2-PEt all show promising results as biomarkers to predict therapy response and outcome, for eventual integration into clinical practice, future studies will have to clarify at what timepoint this integration has to optimally take place.
Collapse
Affiliation(s)
- Jasper J. L. van Geel
- Department of Medical Oncology, University
Medical Center Groningen, University of Groningen, Groningen, The
Netherlands
| | - Erik F. J. de Vries
- Department of Nuclear Medicine and Molecular
Imaging, University Medical Center Groningen, University of Groningen,
Groningen, The Netherlands
| | - Michel van Kruchten
- Department of Medical Oncology, University
Medical Center Groningen, University of Groningen, Groningen, The
Netherlands
| | - Geke A. P. Hospers
- Department of Medical Oncology, University
Medical Center Groningen, University of Groningen, Groningen, The
Netherlands
| | - Andor W. J. M. Glaudemans
- Department of Nuclear Medicine and Molecular
Imaging, University Medical Center Groningen, University of Groningen,
Groningen, The Netherlands
| | - Carolina P. Schröder
- Department of Medical Oncology, University
Medical Center Groningen, University of Groningen, Groningen, The
Netherlands
- Department of Medical Oncology, Netherlands
Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| |
Collapse
|
19
|
Li MP, Liu WC, Sun BL, Zhong NS, Liu ZL, Huang SH, Zhang ZH, Liu JM. Prediction of bone metastasis in non-small cell lung cancer based on machine learning. Front Oncol 2023; 12:1054300. [PMID: 36698411 PMCID: PMC9869148 DOI: 10.3389/fonc.2022.1054300] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/21/2022] [Indexed: 01/12/2023] Open
Abstract
Objective The purpose of this paper was to develop a machine learning algorithm with good performance in predicting bone metastasis (BM) in non-small cell lung cancer (NSCLC) and establish a simple web predictor based on the algorithm. Methods Patients who diagnosed with NSCLC between 2010 and 2018 in the Surveillance, Epidemiology and End Results (SEER) database were involved. To increase the extensibility of the research, data of patients who first diagnosed with NSCLC at the First Affiliated Hospital of Nanchang University between January 2007 and December 2016 were also included in this study. Independent risk factors for BM in NSCLC were screened by univariate and multivariate logistic regression. At this basis, we chose six commonly machine learning algorithms to build predictive models, including Logistic Regression (LR), Decision tree (DT), Random Forest (RF), Gradient Boosting Machine (GBM), Naive Bayes classifiers (NBC) and eXtreme gradient boosting (XGB). Then, the best model was identified to build the web-predictor for predicting BM of NSCLC patients. Finally, area under receiver operating characteristic curve (AUC), accuracy, sensitivity and specificity were used to evaluate the performance of these models. Results A total of 50581 NSCLC patients were included in this study, and 5087(10.06%) of them developed BM. The sex, grade, laterality, histology, T stage, N stage, and chemotherapy were independent risk factors for NSCLC. Of these six models, the machine learning model built by the XGB algorithm performed best in both internal and external data setting validation, with AUC scores of 0.808 and 0.841, respectively. Then, the XGB algorithm was used to build a web predictor of BM from NSCLC. Conclusion This study developed a web predictor based XGB algorithm for predicting the risk of BM in NSCLC patients, which may assist doctors for clinical decision making.
Collapse
Affiliation(s)
- Meng-Pan Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China,The First Clinical Medical College of Nanchang University, Nanchang, China
| | - Wen-Cai Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China,The First Clinical Medical College of Nanchang University, Nanchang, China,Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Bo-Lin Sun
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China,Institute of Spine and Spinal Cord, Nanchang University, Nanchang, China
| | - Nan-Shan Zhong
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China,Institute of Spine and Spinal Cord, Nanchang University, Nanchang, China
| | - Zhi-Li Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China,Institute of Spine and Spinal Cord, Nanchang University, Nanchang, China
| | - Shan-Hu Huang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China,Institute of Spine and Spinal Cord, Nanchang University, Nanchang, China
| | - Zhi-Hong Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China,Institute of Spine and Spinal Cord, Nanchang University, Nanchang, China,*Correspondence: Jia-Ming Liu, ; Zhi-Hong Zhang,
| | - Jia-Ming Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China,Institute of Spine and Spinal Cord, Nanchang University, Nanchang, China,*Correspondence: Jia-Ming Liu, ; Zhi-Hong Zhang,
| |
Collapse
|
20
|
Tian A, Lv H, Liu W, Zhao J, Zhao S, Wang K, Song C. Pseudoprogression after advanced first-line endocrine therapy in metastatic breast cancer with bone metastasis: A case report. Front Oncol 2023; 12:1099164. [PMID: 36686812 PMCID: PMC9845761 DOI: 10.3389/fonc.2022.1099164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023] Open
Abstract
Approximately 75% of patients with advanced breast cancer develop bone metastasis, which significantly affects both the quality of life and the survival rate of patients. Accurate determination of the status of bone metastases is important for developing treatment strategies and the prognosis of the disease. Here, we report the case of a 33-year-old patient with advanced metastatic breast cancer (MBC) and multiple bone metastases, in which advanced first-line endocrine therapy and second-line chemotherapy were both considered unsuccessful according to the efficacy evaluation by conventional imaging. Considering the possibility of bone pseudoprogression, the original endocrine scheme was reapplied, and bone metastases achieved a great response of non-complete response (CR)/non-progressive disease (PD). This case showed that, in the course of therapy for the disease, if bone scintigraphy (BS) shows increased lesion density or new lesions, this probably indicates a favorable response (osteoblastic repair of osteolytic lesions) to therapy, and not the worsening of metastatic lesions, called bone pseudoprogression. This paper will provide new insights into strategies for the treatment of bone metastasis and shows the significance of distinguishing osteoblastic bone repair from real bone lesion progression in clinical settings.
Collapse
Affiliation(s)
- Aijuan Tian
- Department of Nuclear Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Huiyun Lv
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Wei Liu
- Department of Radiology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Jinbo Zhao
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Shanshan Zhao
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Kainan Wang
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Chen Song
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China,*Correspondence: Chen Song,
| |
Collapse
|
21
|
Makhlin I, Korhonen KE, Martin ML, Gillman J, Schubert E, Pantel AR, Mankoff DA, Clark AS. 18F-FDG PET/CT for the Evaluation of Therapy Response in Hormone Receptor-Positive Bone-Dominant Metastatic Breast Cancer. Radiol Imaging Cancer 2022; 4:e220032. [PMID: 36269154 PMCID: PMC9713595 DOI: 10.1148/rycan.220032] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 08/06/2022] [Accepted: 09/13/2022] [Indexed: 05/31/2023]
Abstract
Fluorine 18 (18F) fluorodeoxyglucose (FDG) PET/CT has shown promise for use in assessing treatment response in patients with bone-only or bone-dominant (BD) metastatic breast cancer (mBC). In this single-institution, prospective single-arm study of 23 women (median age, 59 years [range, 38-81 years]) with biopsy-proven estrogen receptor-positive bone-only or BD mBC about to begin new endocrine therapy between October 3, 2013, and August 3, 2018, the value of early 4-week 18F-FDG PET/CT in predicting progression-free survival (PFS) was evaluated. 18F-FDG PET/CT was performed at baseline, 4 weeks, and 12 weeks. Maximum standardized uptake value (SUVmax) and peak SUV (SUVpeak) were measured for up to five index lesions. The primary end point was PFS. Secondary end points were overall survival (OS) and time to skeletal-related events (tSREs). All end points were compared between responders (reduction of 30% or more in the sum of SUVmax for target lesions) and nonresponders at 4 weeks and 12 weeks. Percentage change from baseline in SUVmax at 4- and 12-week 18F-FDG PET/CT were highly correlated (r = 0.81). At the 4-week time point PET responders had numerically longer PFS (14.2 months vs 6.3 months; P = .53), OS (44.0 months vs 29.7 months; P = .47), and tSRE (27.4 months vs 25.2 months; P = .66) compared with nonresponders, suggesting the clinical utility of 4-week 18F-FDG PET/CT as an early predictor of treatment failure. Keywords: Breast Cancer, Metastatic Breast Cancer, Bone-Dominant Metastatic Breast Cancer, FDG PET/CT, Estrogen-Receptor Positive Metastatic Breast Cancer Supplemental material is available for this article. Clinical trial registration no. NCT04316117 © RSNA, 2022.
Collapse
Affiliation(s)
| | | | - Melissa Lynne Martin
- From the Department of Medicine, Division of Hematology &
Oncology (I.M., A.S.C.), Department of Radiology, Division of Nuclear Medicine
Imaging and Therapy (K.E.K., E.S., A.R.P., D.A.M.), Department of Biostatistics,
Epidemiology, and Informatics (M.L.M.), Department of Radiology (J.G.), and
Abramson Cancer Center (D.A.M., A.S.C.), University of Pennsylvania, 3400 Civic
Center Blvd, South Tower, #10-156, Philadelphia, PA 19104
| | - Jennifer Gillman
- From the Department of Medicine, Division of Hematology &
Oncology (I.M., A.S.C.), Department of Radiology, Division of Nuclear Medicine
Imaging and Therapy (K.E.K., E.S., A.R.P., D.A.M.), Department of Biostatistics,
Epidemiology, and Informatics (M.L.M.), Department of Radiology (J.G.), and
Abramson Cancer Center (D.A.M., A.S.C.), University of Pennsylvania, 3400 Civic
Center Blvd, South Tower, #10-156, Philadelphia, PA 19104
| | - Erin Schubert
- From the Department of Medicine, Division of Hematology &
Oncology (I.M., A.S.C.), Department of Radiology, Division of Nuclear Medicine
Imaging and Therapy (K.E.K., E.S., A.R.P., D.A.M.), Department of Biostatistics,
Epidemiology, and Informatics (M.L.M.), Department of Radiology (J.G.), and
Abramson Cancer Center (D.A.M., A.S.C.), University of Pennsylvania, 3400 Civic
Center Blvd, South Tower, #10-156, Philadelphia, PA 19104
| | - Austin Ryan Pantel
- From the Department of Medicine, Division of Hematology &
Oncology (I.M., A.S.C.), Department of Radiology, Division of Nuclear Medicine
Imaging and Therapy (K.E.K., E.S., A.R.P., D.A.M.), Department of Biostatistics,
Epidemiology, and Informatics (M.L.M.), Department of Radiology (J.G.), and
Abramson Cancer Center (D.A.M., A.S.C.), University of Pennsylvania, 3400 Civic
Center Blvd, South Tower, #10-156, Philadelphia, PA 19104
| | - David A. Mankoff
- From the Department of Medicine, Division of Hematology &
Oncology (I.M., A.S.C.), Department of Radiology, Division of Nuclear Medicine
Imaging and Therapy (K.E.K., E.S., A.R.P., D.A.M.), Department of Biostatistics,
Epidemiology, and Informatics (M.L.M.), Department of Radiology (J.G.), and
Abramson Cancer Center (D.A.M., A.S.C.), University of Pennsylvania, 3400 Civic
Center Blvd, South Tower, #10-156, Philadelphia, PA 19104
| | - Amy S. Clark
- From the Department of Medicine, Division of Hematology &
Oncology (I.M., A.S.C.), Department of Radiology, Division of Nuclear Medicine
Imaging and Therapy (K.E.K., E.S., A.R.P., D.A.M.), Department of Biostatistics,
Epidemiology, and Informatics (M.L.M.), Department of Radiology (J.G.), and
Abramson Cancer Center (D.A.M., A.S.C.), University of Pennsylvania, 3400 Civic
Center Blvd, South Tower, #10-156, Philadelphia, PA 19104
| |
Collapse
|
22
|
Cook GJR. Imaging of Bone Metastases in Breast Cancer. Semin Nucl Med 2022; 52:531-541. [PMID: 35236615 PMCID: PMC7616189 DOI: 10.1053/j.semnuclmed.2022.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 11/11/2022]
Abstract
Bone metastases are a common site of spread in advanced breast cancer and responsible for morbidity and high health care costs. Imaging contributes to staging and response assessment of the skeleton and has been instrumental in guiding patient management for several decades. Historically this has been with radiographs, computed tomography and bone scans. More recently, molecular and hybrid imaging methods have undergone significant development, including the addition of single photon emission computed tomography/computed tomography to the bone scan, positron emission tomography, with bone-specific and tumor-specific tracers, and magnetic resonance imaging with complementary functional diffusion-weighted imaging. These have allowed different aspects of the abnormal biology associated with bone metastases to be explored. There is ability to interrogate the bone microenvironment with bone-specific tracers and cancer cell characteristics with tumor-specific methods that complement morphological appearances on computed tomography or magnetic resonance imaging. Alongside the advent of novel, more effective and nuanced therapies for bone metastases in breast cancer, there is accumulating evidence that the developments in imaging allow more sensitive and specific detection of bone metastases as well as more accurate and earlier assessment of treatment response leading to improvements in patient management.
Collapse
Affiliation(s)
- Gary J R Cook
- Cancer Imaging Department, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK; King's College London & Guy's and St Thomas' PET Centre, St Thomas' Hospital, London, UK.
| |
Collapse
|
23
|
Kairemo K, Macapinlac HA. Oncology, bone metastases. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00032-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
24
|
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
|
25
|
Yao YB, Zheng XE, Luo XB, Wu AM. Incidence, prognosis and nomograms of breast cancer with bone metastases at initial diagnosis: a large population-based study. Am J Transl Res 2021; 13:10248-10261. [PMID: 34650694 PMCID: PMC8507056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 06/13/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Bone is the most common metastatic site for breast cancer, and patients' condition will deteriorate when it occurs. METHODS We performed a retrospective analysis on 6482 breast cancer patients with bone metastases (BCBM), who were selected from the Surveillance, Epidemiology, and End Result (SEER) 18 registry database. The optimal age cut-points were generated by using the X-tile software. By using Cox regression, we selected independent prognostic factors from 21 variables, and plotted a visual nomogram to predict the probability of surviving to the median survival time. We also diagrammed a competing risk nomogram on the basis of competitive risk model. RESULTS Compared with other three common metastatic sites, the incidence of bone metastasis was the highest for patients with breast cancer. The incidence of BCBM peaked around the age of 60, and a large majority of patients were between the ages of 50 and 70. The survival rate decreased with age, and the median survival time was about 19 months. Factors of age, race, marital status, grade, human epidermal growth factor receptor-2 (HER2) receptor, hormone receptor, concurrent brain metastasis, concurrent liver metastasis, concurrent lung metastasis, surgery and chemotherapy are strongly related to the prognosis of patients with BCBM. It was revealed that the C-index of the nomogram was 0.72 and the calibration curves showed good agreement between the nomogram prediction and actual observation. CONCLUSION Our practical nomograms provide a visual and user-friendly tool in the risk evaluation and prognostic prediction for breast cancer patients with bone metastases.
Collapse
Affiliation(s)
- Yu-Bin Yao
- The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, Zhejiang, China
| | - Xue-Er Zheng
- The First Clinical Medical College of Zhejiang Chinese Medical UniversityHangzhou 310053, Zhejiang, China
| | - Xiao-Bin Luo
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhou 325027, China
| | - Ai-Min Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou Medical UniversityWenzhou 325027, China
| |
Collapse
|
26
|
Meikle SR, Sossi V, Roncali E, Cherry SR, Banati R, Mankoff D, Jones T, James M, Sutcliffe J, Ouyang J, Petibon Y, Ma C, El Fakhri G, Surti S, Karp JS, Badawi RD, Yamaya T, Akamatsu G, Schramm G, Rezaei A, Nuyts J, Fulton R, Kyme A, Lois C, Sari H, Price J, Boellaard R, Jeraj R, Bailey DL, Eslick E, Willowson KP, Dutta J. Quantitative PET in the 2020s: a roadmap. Phys Med Biol 2021; 66:06RM01. [PMID: 33339012 PMCID: PMC9358699 DOI: 10.1088/1361-6560/abd4f7] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Positron emission tomography (PET) plays an increasingly important role in research and clinical applications, catalysed by remarkable technical advances and a growing appreciation of the need for reliable, sensitive biomarkers of human function in health and disease. Over the last 30 years, a large amount of the physics and engineering effort in PET has been motivated by the dominant clinical application during that period, oncology. This has led to important developments such as PET/CT, whole-body PET, 3D PET, accelerated statistical image reconstruction, and time-of-flight PET. Despite impressive improvements in image quality as a result of these advances, the emphasis on static, semi-quantitative 'hot spot' imaging for oncologic applications has meant that the capability of PET to quantify biologically relevant parameters based on tracer kinetics has not been fully exploited. More recent advances, such as PET/MR and total-body PET, have opened up the ability to address a vast range of new research questions, from which a future expansion of applications and radiotracers appears highly likely. Many of these new applications and tracers will, at least initially, require quantitative analyses that more fully exploit the exquisite sensitivity of PET and the tracer principle on which it is based. It is also expected that they will require more sophisticated quantitative analysis methods than those that are currently available. At the same time, artificial intelligence is revolutionizing data analysis and impacting the relationship between the statistical quality of the acquired data and the information we can extract from the data. In this roadmap, leaders of the key sub-disciplines of the field identify the challenges and opportunities to be addressed over the next ten years that will enable PET to realise its full quantitative potential, initially in research laboratories and, ultimately, in clinical practice.
Collapse
Affiliation(s)
- Steven R Meikle
- Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
- Brain and Mind Centre, The University of Sydney, Australia
| | - Vesna Sossi
- Department of Physics and Astronomy, University of British Columbia, Canada
| | - Emilie Roncali
- Department of Biomedical Engineering, University of California, Davis, United States of America
| | - Simon R Cherry
- Department of Biomedical Engineering, University of California, Davis, United States of America
- Department of Radiology, University of California, Davis, United States of America
| | - Richard Banati
- Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
- Brain and Mind Centre, The University of Sydney, Australia
- Australian Nuclear Science and Technology Organisation, Sydney, Australia
| | - David Mankoff
- Department of Radiology, University of Pennsylvania, United States of America
| | - Terry Jones
- Department of Radiology, University of California, Davis, United States of America
| | - Michelle James
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), CA, United States of America
- Department of Neurology and Neurological Sciences, Stanford University, CA, United States of America
| | - Julie Sutcliffe
- Department of Biomedical Engineering, University of California, Davis, United States of America
- Department of Internal Medicine, University of California, Davis, CA, United States of America
| | - Jinsong Ouyang
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Yoann Petibon
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Chao Ma
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Suleman Surti
- Department of Radiology, University of Pennsylvania, United States of America
| | - Joel S Karp
- Department of Radiology, University of Pennsylvania, United States of America
| | - Ramsey D Badawi
- Department of Biomedical Engineering, University of California, Davis, United States of America
- Department of Radiology, University of California, Davis, United States of America
| | - Taiga Yamaya
- National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Go Akamatsu
- National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Georg Schramm
- Department of Imaging and Pathology, Nuclear Medicine & Molecular imaging, KU Leuven, Belgium
| | - Ahmadreza Rezaei
- Department of Imaging and Pathology, Nuclear Medicine & Molecular imaging, KU Leuven, Belgium
| | - Johan Nuyts
- Department of Imaging and Pathology, Nuclear Medicine & Molecular imaging, KU Leuven, Belgium
| | - Roger Fulton
- Brain and Mind Centre, The University of Sydney, Australia
- Department of Medical Physics, Westmead Hospital, Sydney, Australia
| | - André Kyme
- Brain and Mind Centre, The University of Sydney, Australia
- School of Biomedical Engineering, Faculty of Engineering and IT, The University of Sydney, Australia
| | - Cristina Lois
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Hasan Sari
- Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
- Athinoula A. Martinos Center, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
| | - Julie Price
- Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
- Athinoula A. Martinos Center, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
| | - Ronald Boellaard
- Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, location VUMC, Netherlands
| | - Robert Jeraj
- Departments of Medical Physics, Human Oncology and Radiology, University of Wisconsin, United States of America
- Faculty of Mathematics and Physics, University of Ljubljana, Slovenia
| | - Dale L Bailey
- Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
- Faculty of Science, The University of Sydney, Australia
| | - Enid Eslick
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
| | - Kathy P Willowson
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
- Faculty of Science, The University of Sydney, Australia
| | - Joyita Dutta
- Department of Electrical and Computer Engineering, University of Massachusetts Lowell, United States of America
| |
Collapse
|
27
|
Abstract
Response evaluation for cancer treatment consists primarily of clinical and radiological assessments. In addition, a limited number of serum biomarkers that assess treatment response are available for a small subset of malignancies. Through recent technological innovations, new methods for measuring tumor burden and treatment response are becoming available. By utilization of highly sensitive techniques, tumor-specific mutations in circulating DNA can be detected and circulating tumor DNA (ctDNA) can be quantified. These so-called liquid biopsies provide both molecular information about the genomic composition of the tumor and opportunities to evaluate tumor response during therapy. Quantification of tumor-specific mutations in plasma correlates well with tumor burden. Moreover, with liquid biopsies, it is also possible to detect mutations causing secondary resistance during treatment. This review focuses on the clinical utility of ctDNA as a response and follow-up marker in patients with non-small cell lung cancer, melanoma, colorectal cancer, and breast cancer. Relevant studies were retrieved from a literature search using PubMed database. An overview of the available literature is provided and the relevance of ctDNA as a response marker in anti-cancer therapy for clinical practice is discussed. We conclude that the use of plasma-derived ctDNA is a promising tool for treatment decision-making based on predictive testing, detection of resistance mechanisms, and monitoring tumor response. Necessary steps for translation to daily practice and future perspectives are discussed.
Collapse
|
28
|
Clézardin P, Coleman R, Puppo M, Ottewell P, Bonnelye E, Paycha F, Confavreux CB, Holen I. Bone metastasis: mechanisms, therapies, and biomarkers. Physiol Rev 2020; 101:797-855. [PMID: 33356915 DOI: 10.1152/physrev.00012.2019] [Citation(s) in RCA: 176] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Skeletal metastases are frequent complications of many cancers, causing bone complications (fractures, bone pain, disability) that negatively affect the patient's quality of life. Here, we first discuss the burden of skeletal complications in cancer bone metastasis. We then describe the pathophysiology of bone metastasis. Bone metastasis is a multistage process: long before the development of clinically detectable metastases, circulating tumor cells settle and enter a dormant state in normal vascular and endosteal niches present in the bone marrow, which provide immediate attachment and shelter, and only become active years later as they proliferate and alter the functions of bone-resorbing (osteoclasts) and bone-forming (osteoblasts) cells, promoting skeletal destruction. The molecular mechanisms involved in mediating each of these steps are described, and we also explain how tumor cells interact with a myriad of interconnected cell populations in the bone marrow, including a rich vascular network, immune cells, adipocytes, and nerves. We discuss metabolic programs that tumor cells could engage with to specifically grow in bone. We also describe the progress and future directions of existing bone-targeted agents and report emerging therapies that have arisen from recent advances in our understanding of the pathophysiology of bone metastases. Finally, we discuss the value of bone turnover biomarkers in detection and monitoring of progression and therapeutic effects in patients with bone metastasis.
Collapse
Affiliation(s)
- Philippe Clézardin
- INSERM, Research Unit UMR_S1033, LyOS, Faculty of Medicine Lyon-Est, University of Lyon 1, Lyon, France.,Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Rob Coleman
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Margherita Puppo
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Penelope Ottewell
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Edith Bonnelye
- INSERM, Research Unit UMR_S1033, LyOS, Faculty of Medicine Lyon-Est, University of Lyon 1, Lyon, France
| | - Frédéric Paycha
- Service de Médecine Nucléaire, Hôpital Lariboisière, Paris, France
| | - Cyrille B Confavreux
- INSERM, Research Unit UMR_S1033, LyOS, Faculty of Medicine Lyon-Est, University of Lyon 1, Lyon, France.,Service de Rhumatologie Sud, CEMOS-Centre Expert des Métastases Osseuses, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Ingunn Holen
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
29
|
Gillman JA, Pantel AR, Mankoff DA, Edmonds CE. Update on Quantitative Imaging for Predicting and Assessing Response in Oncology. Semin Nucl Med 2020; 50:505-517. [PMID: 33059820 PMCID: PMC9788668 DOI: 10.1053/j.semnuclmed.2020.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Molecular imaging has revolutionized clinical oncology by imaging-specific facets of cancer biology. Through noninvasive measurements of tumor physiology, targeted radiotracers can serve as biomarkers for disease characterization, prognosis, response assessment, and predicting long-term response/survival. In turn, these imaging biomarkers can be utilized to tailor therapeutic regimens to tumor biology. In this article, we review biomarker applications for response assessment and predicting long-term outcomes. 18F-fluorodeoxyglucose (FDG), a measure of cellular glucose metabolism, is discussed in the context of lymphoma and breast and lung cancer. FDG has gained widespread clinical acceptance and has been integrated into the routine clinical care of several malignancies, most notably lymphoma. The novel radiotracers 16α-18F-fluoro-17β-estradiol and 18F-fluorothymidine are reviewed in application to the early prediction of response assessment of breast cancer. Through illustrative examples, we explore current and future applications of molecular imaging biomarkers in the advancement of precision medicine.
Collapse
Affiliation(s)
- Jennifer A Gillman
- Department of Radiology, Division of Nuclear Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Austin R Pantel
- Department of Radiology, Division of Nuclear Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - David A Mankoff
- Department of Radiology, Division of Nuclear Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Christine E Edmonds
- Department of Radiology, Division of Nuclear Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA.
| |
Collapse
|
30
|
van Es SC, Velleman T, Elias SG, Bensch F, Brouwers AH, Glaudemans AWJM, Kwee TC, Iersel MWV, Maduro JH, Oosting SF, de Vries EGE, Schröder CP. Assessment of Bone Lesions with 18F-FDG PET Compared with 99mTc Bone Scintigraphy Leads to Clinically Relevant Differences in Metastatic Breast Cancer Management. J Nucl Med 2020; 62:177-183. [PMID: 32817140 DOI: 10.2967/jnumed.120.244640] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/07/2020] [Indexed: 11/16/2022] Open
Abstract
It is unknown whether assessment of potential bone lesions in metastatic breast cancer (MBC) by 18F-FDG PET instead of 99mTc bone scintigraphy (BS) supports clinically relevant changes in MBC management. Therefore, we retrospectively compared management recommendations based on bone lesion assessment by 18F-FDG PET plus contrast-enhanced CT (ceCT) or BS plus ceCT, for patients with newly diagnosed MBC. Methods: Baseline ceCT, BS, and 18F-FDG PET for all patients included in the IMPACT-MBC study (NCT01957332) at the University Medical Center Groningen were reviewed for bone lesions. If bone lesions were found by any imaging modality, virtual MBC management recommendations were made by a multidisciplinary expert panel, based on either 18F-FDG PET plus ceCT or BS plus ceCT. The panel had access to standard clinicopathologic information and baseline imaging findings outside the skeleton. Clinically relevant management differences between the 2 recommendations were defined either as different treatment intent (curative, noncurative, or unable to determine) or as different systemic or local treatment. If no bone lesions were found by any imaging modality, the patients were included in the analyses without expert review. Results: In total, 3,473 unequivocal bone lesions were identified in 102 evaluated patients (39% by ceCT, 26% by BS, and 87% by 18F-FDG PET). Additional bone lesions on 18F-FDG PET plus ceCT compared with BS plus ceCT led to change in MBC management recommendations in 16% of patients (95% CI, 10%-24%). BS also changed management compared with 18F-FDG PET in 1 patient (1%; 95% CI, 0%-5%). In 26% (95% CI, 19%-36%) of patients, an additional 18F-FDG PET exam was requested, because BS provided insufficient information. Conclusion: In this exploratory analysis of newly diagnosed MBC patients, 18F-FDG PET versus BS to assess bone lesions resulted in clinically relevant management differences in 16% of patients. BS delivered insufficient information in over one fourth of patients, resulting in an additional request for 18F-FDG PET. On the basis of these data, 18F-FDG PET should be considered a primary imaging modality for assessment of bone lesions in newly diagnosed MBC.
Collapse
Affiliation(s)
- Suzanne C van Es
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ton Velleman
- Department of Radiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sjoerd G Elias
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Frederike Bensch
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Adrienne H Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and
| | - Andor W J M Glaudemans
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and
| | - Thomas C Kwee
- Department of Radiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marleen Woltman-van Iersel
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - John H Maduro
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sjoukje F Oosting
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Carolina P Schröder
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
31
|
Ulaner GA, Jhaveri K, Chandarlapaty S, Hatzoglou V, Riedl CC, Lewis JS, Mauguen A. Head-to-Head Evaluation of 18F-FES and 18F-FDG PET/CT in Metastatic Invasive Lobular Breast Cancer. J Nucl Med 2020; 62:326-331. [PMID: 32680923 DOI: 10.2967/jnumed.120.247882] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/15/2020] [Indexed: 02/07/2023] Open
Abstract
Invasive lobular carcinoma (ILC) demonstrates lower conspicuity on 18F-FDG PET than the more common invasive ductal carcinoma. Other molecular imaging methods may be needed for evaluation of this malignancy. As ILC is nearly always (95%) estrogen receptor (ER)-positive, ER-targeting PET tracers such as 16α-18F-fluoroestradiol (18F-FES) may have value. We reviewed prospective trials at Memorial Sloan Kettering Cancer Center using 18F-FES PET/CT to evaluate metastatic ILC patients with synchronous 18F-FDG and 18F-FES PET/CT imaging, which allowed a head-to-head comparison of these 2 PET tracers. Methods: Six prospective clinical trials using 18F-FES PET/CT in patients with metastatic breast cancer were performed at Memorial Sloan Kettering Cancer Center from 2008 to 2019. These trials included 92 patients, of whom 14 (15%) were of ILC histology. Seven of 14 patients with ILC had 18F-FDG PET/CT performed within 5 wk of the research 18F-FES PET/CT and no intervening change in management. For these 7 patients, the 18F-FES and 18F-FDG PET/CT studies were analyzed to determine the total number of tracer-avid lesions, organ systems of involvement, and SUVmax of each organ system for both tracers. Results: In the 7 comparable pairs of scans, there were a total of 254 18F-FES-avid lesions (SUVmax, 2.6-17.9) and 111 18F-FDG-avid lesions (SUVmax, 3.3-9.9) suggestive of malignancy. For 5 of 7 (71%) ILC patients, 18F-FES PET/CT detected more metastatic lesions than 18F-FDG PET/CT. In the same 5 of 7 patients, the SUVmax of 18F-FES-avid lesions was greater than the SUVmax of 18F-FDG-avid lesions. One patient had 18F-FES-avid metastases with no corresponding 18F-FDG-avid metastases. There were no patients with 18F-FDG-avid distant metastases without 18F-FES-avid distant metastases, although in one patient liver metastases were evident on 18F-FDG but not on 18F-FES PET. Conclusion: 18F-FES PET/CT compared favorably with 18F-FDG PET/CT for detection of metastases in patients with metastatic ILC. Larger prospective trials of 18F-FES PET/CT in ILC should be considered to evaluate ER-targeted imaging for clinical value in patients with this histology of breast cancer.
Collapse
Affiliation(s)
- Gary A Ulaner
- Molecular Imaging and Therapy, Hoag Family Cancer Institute, Newport Beach, California .,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Komal Jhaveri
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Sarat Chandarlapaty
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Vaios Hatzoglou
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill Cornell Medical College, New York, New York; and
| | - Christopher C Riedl
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill Cornell Medical College, New York, New York; and
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill Cornell Medical College, New York, New York; and
| | - Audrey Mauguen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
32
|
Boers J, de Vries EFJ, Glaudemans AWJM, Hospers GAP, Schröder CP. Application of PET Tracers in Molecular Imaging for Breast Cancer. Curr Oncol Rep 2020; 22:85. [PMID: 32627087 PMCID: PMC7335757 DOI: 10.1007/s11912-020-00940-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Molecular imaging with positron emission tomography (PET) is a powerful tool to visualize breast cancer characteristics. Nonetheless, implementation of PET imaging into cancer care is challenging, and essential steps have been outlined in the international "imaging biomarker roadmap." In this review, we identify hurdles and provide recommendations for implementation of PET biomarkers in breast cancer care, focusing on the PET tracers 2-[18F]-fluoro-2-deoxyglucose ([18F]-FDG), sodium [18F]-fluoride ([18F]-NaF), 16α-[18F]-fluoroestradiol ([18F]-FES), and [89Zr]-trastuzumab. RECENT FINDINGS Technical validity of [18F]-FDG, [18F]-NaF, and [18F]-FES is established and supported by international guidelines. However, support for clinical validity and utility is still pending for these PET tracers in breast cancer, due to variable endpoints and procedures in clinical studies. Assessment of clinical validity and utility is essential towards implementation; however, these steps are still lacking for PET biomarkers in breast cancer. This could be solved by adding PET biomarkers to randomized trials, development of imaging data warehouses, and harmonization of endpoints and procedures.
Collapse
Affiliation(s)
- Jorianne Boers
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, The Netherlands
| | - Erik F J de Vries
- Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Andor W J M Glaudemans
- Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Geke A P Hospers
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, The Netherlands
| | - Carolina P Schröder
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, The Netherlands.
| |
Collapse
|
33
|
Peterson LM, Kurland BF, Yan F, Jiresova AN, Gadi VK, Specht JM, Gralow JR, Schubert EK, Link JM, Krohn KA, Eary JF, Mankoff DA, Linden HM. 18F-Fluoroestradiol PET Imaging in a Phase II Trial of Vorinostat to Restore Endocrine Sensitivity in ER+/HER2- Metastatic Breast Cancer. J Nucl Med 2020; 62:184-190. [PMID: 32591490 DOI: 10.2967/jnumed.120.244459] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/27/2020] [Indexed: 12/23/2022] Open
Abstract
Histone deacetylase inhibitors (HDACIs) may overcome endocrine resistance in estrogen receptor-positive (ER+) metastatic breast cancer. We tested whether 18F-fluoroestradiol PET imaging would elucidate the pharmacodynamics of combination HDACIs and endocrine therapy. Methods: Patients with ER+/human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer with prior clinical benefit from endocrine therapy but later progression on aromatase inhibitor (AI) therapy were given vorinostat (400 mg daily) sequentially or simultaneously with AI. 18F-fluoroestradiol PET and 18F-FDG PET scans were performed at baseline, week 2, and week 8. Results: Eight patients were treated sequentially, and then 15 simultaneously. Eight patients had stable disease at week 8, and 6 of these 8 patients had more than 6 mo of stable disease. Higher baseline 18F-fluoroestradiol uptake was associated with longer progression-free survival. 18F-fluoroestradiol uptake did not systematically increase with vorinostat exposure, indicating no change in regional ER estradiol binding, and 18F-FDG uptake did not show a significant decrease, as would have been expected with tumor regression. Conclusion: Simultaneous HDACIs and AI dosing in patients with cancer resistant to AI alone showed clinical benefit (6 or more months without progression) in 4 of 10 evaluable patients. Higher 18F-fluoroestradiol PET uptake identified patients likely to benefit from combination therapy, but vorinostat did not change ER expression at the level of detection of 18F-fluoroestradiol PET.
Collapse
Affiliation(s)
- Lanell M Peterson
- Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, Seattle, Washington
| | - Brenda F Kurland
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Fengting Yan
- Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, Seattle, Washington
| | - Alena Novakova- Jiresova
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Vijayakrishna K Gadi
- Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, Seattle, Washington.,Clinical Research and Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Jennifer M Specht
- Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, Seattle, Washington
| | - Julie R Gralow
- Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, Seattle, Washington
| | - Erin K Schubert
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jeanne M Link
- Department of Diagnostic Radiology, Oregon Health and Science University, Portland, Oregon; and
| | - Kenneth A Krohn
- Department of Diagnostic Radiology, Oregon Health and Science University, Portland, Oregon; and
| | - Janet F Eary
- Cancer Imaging Program, National Cancer Institute, Bethesda, Maryland
| | - David A Mankoff
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hannah M Linden
- Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, Seattle, Washington
| |
Collapse
|
34
|
Cook GJR, Goh V. Molecular Imaging of Bone Metastases and Their Response to Therapy. J Nucl Med 2020; 61:799-806. [PMID: 32245899 DOI: 10.2967/jnumed.119.234260] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 03/30/2020] [Indexed: 12/18/2022] Open
Abstract
Bone metastases are common, especially in more prevalent malignancies such as breast and prostate cancer. They cause significant morbidity and draw on health-care resources. Molecular and hybrid imaging techniques, including SPECT/CT, PET/CT, and whole-body MRI with diffusion-weighted imaging, have improved diagnostic accuracy in staging the skeleton compared with previous standard imaging methods, allowing earlier tailored treatment. With the introduction of several effective treatment options, it is now even more important to detect and monitor response in bone metastases accurately. Conventional imaging, including radiographs, CT, MRI, and bone scintigraphy, are recognized as being insensitive and nonspecific for response monitoring in a clinically relevant time frame. Early reports of molecular and hybrid imaging techniques, as well as whole-body MRI, promise an earlier and more accurate prediction of response versus nonresponse but have yet to be adopted routinely in clinical practice. We summarize the role of new molecular and hybrid imaging methods, including SPECT/CT, PET/CT, and whole-body MRI. These modalities are associated with improvements in diagnostic accuracy for the staging and response assessment of skeletal metastases over standard imaging methods, being able to quantify biologic processes related to the bone microenvironment as well as tumor cells. The described improvements in the imaging of bone metastases and their response to therapy have led to adoption of some of these methods into routine clinical practice in some centers. These methods also provide a better way to assess the treatment response of bone metastases in clinical trials.
Collapse
Affiliation(s)
- Gary J R Cook
- Cancer Imaging Department, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Vicky Goh
- Cancer Imaging Department, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| |
Collapse
|
35
|
Taralli S, Caldarella C, Lorusso M, Scolozzi V, Altini C, Rubini G, Calcagni ML. Comparison between 18F-FDG and 18F-NaF PET imaging for assessing bone metastases in breast cancer patients: a literature review. Clin Transl Imaging 2020. [DOI: 10.1007/s40336-020-00363-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
36
|
Ulaner GA, Saura C, Piha-Paul SA, Mayer I, Quinn D, Jhaveri K, Stone B, Shahin S, Mann G, Dujka M, Bryce R, Meric-Bernstam F, Solit DB, Hyman DM. Impact of FDG PET Imaging for Expanding Patient Eligibility and Measuring Treatment Response in a Genome-Driven Basket Trial of the Pan-HER Kinase Inhibitor, Neratinib. Clin Cancer Res 2019; 25:7381-7387. [PMID: 31548342 PMCID: PMC7418635 DOI: 10.1158/1078-0432.ccr-19-1658] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/01/2019] [Accepted: 09/11/2019] [Indexed: 12/14/2022]
Abstract
PURPOSE To determine whether FDG PET can expand eligibility in biomarker-selected clinical trials by providing a means to quantitate response in patients with non-assessable disease by RECIST. EXPERIMENTAL DESIGN SUMMIT (NCT01953926) is a multicenter phase II "basket" trial of the Pan-HER kinase inhibitor, neratinib. Patients had advanced ERBB2 (HER2)-mutant solid tumors, ≥1 measurable lesion, preferably defined unidimensionally by RECIST v1.1, or alternatively metabolically by PET Response Criteria (PRC). The primary aim was to determine the proportion of additional breast cancer patients accrued using PRC who would have otherwise been ineligible based on RECIST criteria alone. The secondary aim was to determine the concordance of response versus non-response between RECIST and PRC. RESULTS Eighty-one patients with HER2-mutant metastatic breast cancer were accrued; 77 were evaluable for response by RECIST and/or PRC. 63 (82%) were RECIST-evaluable and 14 (18%) were accrued using PRC alone. Bone-only disease (n = 11; 79%) was the most common reason for classification as non-measurable by RECIST. Twenty-nine patients were accrued and followed using both criteria, of which 25 (86%; 95% confidence interval, 68%-96%) were concordant for response versus non-response as defined by RECIST and PRC. CONCLUSIONS PRC allowed patients with non-RECIST measurable disease access to therapy and facilitated more rapid accrual of patients to this trial of a rare biomarker. PRC and RECIST both provided methods of response assessment and were generally concordant. Thus, PRC was useful as a supplement to RECIST criteria. This provides a rationale for including FDG PET measurements in future clinical trials involving rare tumors or rare genomically defined subpopulations of more common cancers.
Collapse
Affiliation(s)
- Gary A Ulaner
- Memorial Sloan Kettering Cancer Center, New York, New York.
- Weill Cornell Medical College, New York, New York
| | - Cristina Saura
- Hospital Universitario Vall d'Hebron, Vall d'Hebrón Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Ingrid Mayer
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - David Quinn
- USC Norris Comprehensive Cancer Center, Los Angeles, California
| | - Komal Jhaveri
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ben Stone
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Grace Mann
- Puma Biotechnology, Los Angeles, California
| | | | | | | | - David B Solit
- Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
| | - David M Hyman
- Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
| |
Collapse
|
37
|
Mankoff DA, Pantel AR, Viswanath V, Karp JS. Advances in PET Diagnostics for Guiding Targeted Cancer Therapy and Studying In Vivo Cancer Biology. CURRENT PATHOBIOLOGY REPORTS 2019; 7:97-108. [PMID: 37092138 PMCID: PMC10117535 DOI: 10.1007/s40139-019-00202-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Purpose of the Review We present an overview of recent advances in positron emission tomography (PET) diagnostics as applied to the study of cancer, specifically as a tool to study in vivo cancer biology and to direct targeted cancer therapy. The review is directed to translational and clinical cancer investigators who may not be familiar with these applications of PET cancer diagnostics, but whose research might benefit from these advancing tools. Recent Findings We highlight recent advances in 3 areas: (1) the translation of PET imaging cancer biomarkers to clinical trials; (2) methods for measuring cancer metabolism in vivo in patients; and (3) advances in PET instrumentation, including total-body PET, that enable new methodologies. We emphasize approaches that have been translated to human studies. Summary PET imaging methodology enables unique in vivo cancer diagnostics that go beyond cancer detection and staging, providing an improved ability to guide cancer treatment and an increased understanding of in vivo human cancer biology.
Collapse
Affiliation(s)
- David A Mankoff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Austin R Pantel
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Varsha Viswanath
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Joel S Karp
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
38
|
Castello A, Lopci E. Response assessment of bone metastatic disease: seeing the forest for the trees RECIST, PERCIST, iRECIST, and PCWG-2. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2019; 63:150-158. [PMID: 31286751 DOI: 10.23736/s1824-4785.19.03193-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Tumor response is often used as a surrogate marker for survival practically in all clinical trials. Therefore, robust and valid response criteria during the course of trials are fundamental for the assessment of response to therapy. This aspect, however, becomes particularly challenging when it comes to bone metastases. In the era of targeted therapies and immune-checkpoint inhibitors (ICI), response assessment by morphologic-based criteria cannot detect the real tumor response and, consequently, fail to demonstrate the actual clinical benefit. This review will focus on some of the most common morphologic and metabolic response criteria and their application for bone lesions, highlighting relative strengths and weaknesses as well as potential future methods in the era of target therapies and immunotherapy with ICI.
Collapse
Affiliation(s)
- Angelo Castello
- Nuclear Medicine Unit, Humanitas Clinical and Research Hospital, IRCCS, Rozzano, Milan, Italy
| | - Egesta Lopci
- Nuclear Medicine Unit, Humanitas Clinical and Research Hospital, IRCCS, Rozzano, Milan, Italy -
| |
Collapse
|
39
|
Harmon SA, Mena E, Shih JH, Adler S, McKinney Y, Bergvall E, Mehralivand S, Sowalsky AG, Couvillon A, Madan RA, Gulley JL, Eary J, Mease RC, Pomper MG, Dahut WL, Turkbey B, Lindenberg L, Choyke PL. A comparison of prostate cancer bone metastases on 18F-Sodium Fluoride and Prostate Specific Membrane Antigen ( 18F-PSMA) PET/CT: Discordant uptake in the same lesion. Oncotarget 2018; 9:37676-37688. [PMID: 30701023 PMCID: PMC6340866 DOI: 10.18632/oncotarget.26481] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/04/2018] [Indexed: 12/27/2022] Open
Abstract
Purpose Prostate-Specific Membrane Antigen (PSMA) PET/CT has been introduced as a sensitive method for characterizing metastatic prostate cancer. The purpose of this study is to compare the spatial concordance of 18F-NaF PET/CT and 18F-PSMA-targeted PET/CT within prostate cancer bone metastases. Methods Prostate cancer patients with known bone metastases underwent PSMA-targeted PET/CT (18F-DCFBC or 18F-DCFPyL) and 18F-NaF PET/CT. In pelvic and spinal lesions detected by both radiotracers, regions-of-interest (ROIs) derived by various thresholds of uptake intensity were compared for spatial colocalization. Overlap volume was correlated with uptake characteristics and disease status. Results The study included 149 lesions in 19 patients. Qualitatively, lesions exhibited a heterogeneous range of spatial concordance between PSMA and NaF uptake from completely matched to completely discordant. Quantitatively, overlap volume decreased as a function of tracer intensity. and disease status, where lesions from patients with castration-sensitive disease showed higher spatial concordance while lesions from patients with castration-resistant disease demonstrated more frequent spatial discordance. Conclusion As metastatic prostate cancer progresses from castration-sensitive to castration-resistant, greater discordance is observed between NaF PET and PSMA PET uptake. This may indicate a possible phenotypic shift to tumor growth that is more independent of bone remodeling via osteoblastic formation.
Collapse
Affiliation(s)
- Stephanie A Harmon
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD, USA.,Molecular Imaging Program, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Esther Mena
- Molecular Imaging Program, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Joanna H Shih
- Biometric Research Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Stephen Adler
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD, USA.,Molecular Imaging Program, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Yolanda McKinney
- Molecular Imaging Program, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Ethan Bergvall
- Molecular Imaging Program, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Sherif Mehralivand
- Molecular Imaging Program, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Adam G Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Anna Couvillon
- Genitourinary Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Ravi A Madan
- Genitourinary Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - James L Gulley
- Genitourinary Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Janet Eary
- Cancer Imaging Program, National Cancer Institute, NIH, Rockville, MD, USA
| | - Ronnie C Mease
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - William L Dahut
- Genitourinary Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Baris Turkbey
- Molecular Imaging Program, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Liza Lindenberg
- Molecular Imaging Program, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Peter L Choyke
- Molecular Imaging Program, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|