1
|
van Geel JJL, Moustaquim J, Boers J, Elias SG, Smeets EMM, Knip JJ, Glaudemans AWJM, de Vries EFJ, Hospers GAP, van Kruchten M, Stokkel M, Oprea-Lager DE, Menke-van der Houven van Oordt WC, de Vries EGE, Schröder CP. Intrapatient 16α-[ 18F]Fluoro-17β-Estradiol PET Heterogeneity as a Prognostic Factor for Endocrine Therapy Response and Survival in Patients with Estrogen Receptor-Positive Metastatic Breast Cancer. J Nucl Med 2025; 66:194-200. [PMID: 39848768 DOI: 10.2967/jnumed.124.268984] [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: 10/16/2024] [Accepted: 12/06/2024] [Indexed: 01/25/2025] Open
Abstract
Intrapatient heterogeneity of estrogen receptor (ER) expression on 16α-[18F]fluoro-17β-estradiol ([18F]FES) PET is related to outcome in patients with ER-positive metastatic breast cancer (MBC), but a validated and practical method to support clinical decision-making is lacking. Therefore, the [18F]FES PET heterogeneity score (i.e., percentage of [18F]FES-positive metastases) was validated as a prognostic factor for endocrine therapy response and survival in a large cohort of patients with newly diagnosed MBC. Furthermore, we explored 2 less laborious methods to predict the [18F]FES PET heterogeneity score. Methods: Patients with ER-positive MBC included in the IMPACT-MBC study, who received baseline [18F]FES and [18F]FDG PET and first-line endocrine therapy, were included in this subanalysis. ER homogeneous (100% [18F]FES-positive lesions) and ER heterogeneous (both [18F]FES-positive and [18F]FES-negative lesions) MBC was distinguished by manual segmentation of all lesions on [18F]FES PET and related to progression-free survival (PFS) and overall survival (OS). In addition, the positive predictive value of the visual assessment and the 5-largest-lesions assessment to predict homogeneous MBC in all lesions on [18F]FES PET was determined. Results: From the 102 MBC patients eligible for the present retrospective subanalysis, 46 had ER homogeneous MBC and 56 had ER heterogeneous MBC. Differences were found between ER homogeneous and ER heterogeneous MBC for median PFS (19.8 vs. 15.0 mo; hazard ratio, 0.63; 95% CI, 0.41-0.96; P = 0.03) and median OS (62.5 vs. 34.7 mo; hazard ratio, 0.65; 95% CI, 0.38-1.08; P = 0.09). Twenty-one (38%) of 61 patients with ER homogeneous MBC by visual analysis and 37 (45%) of 83 patients with ER homogeneous MBC by the 5-largest-lesions method had ER heterogeneous MBC by manual segmentation of all lesions on [18F]FES PET (positive predictive value, 0.66 and 0.55, respectively). Conclusion: Patients with ER-positive homogeneous MBC showed a trend toward superior PFS and OS compared with patients with ER heterogeneous MBC. This analysis confirmed and validated the prognostic value of the [18F]FES PET heterogeneity score for endocrine therapy response and survival in a large cohort of MBC patients. The less laborious visual and 5-largest-lesions methods were inferior compared with assessment based on the [18F]FES PET heterogeneity score in all lesions.
Collapse
Affiliation(s)
- Jasper J L van Geel
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jasmine Moustaquim
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jorianne Boers
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sjoerd G Elias
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Esther M M Smeets
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jelijn J Knip
- Department of Medical Oncology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Andor W J M Glaudemans
- Department of Nuclear Medicine and Molecular Imaging, 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
| | - Geke A P Hospers
- Department of Medical Oncology, 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
| | - Marcel Stokkel
- Department of Nuclear Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daniela E Oprea-Lager
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands; and
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | |
Collapse
|
2
|
Shao Q, Zhang N, Pan X, Zhou W, Wang Y, Chen X, Wu J, Zeng X. A Single-Arm Phase II Clinical Trial of Fulvestrant Combined with Neoadjuvant Chemotherapy of ER+/HER2- Locally Advanced Breast Cancer: Integrated Analysis of 18F-FES PET-CT and Metabolites with Treatment Response. Cancer Res Treat 2025; 57:126-139. [PMID: 38993095 PMCID: PMC11729317 DOI: 10.4143/crt.2023.1251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 07/07/2024] [Indexed: 07/13/2024] Open
Abstract
PURPOSE This Phase II trial was objected to evaluate the efficacy and safety of adding fulvestrant to neoadjuvant chemotherapy in patients with estrogen receptor (ER)+/human epidermal growth factor receptor 2 (HER2)- locally advanced breast cancer (LABC). Additionally, the study aimed to investigate the association of 16α-18F-fluoro-17β-fluoroestradiol (18F-FES) positron emission tomography (PET)-computed tomography (CT) and metabolites with efficacy. MATERIALS AND METHODS Fulvestrant and EC-T regimen were given to ER+/HER2- LABC patients before surgery. At baseline, patients received 18F-FES PET-CT scan, and plasma samples were taken for liquid chromatography-mass spectrometry analysis. The primary endpoint was objective response rate (ORR). Secondary endpoints included total pathologic complete response (tpCR) and safety. RESULTS Among the 36 patients enrolled, the ORR was 86.1%, the tpCR rate was 8.3%. The incidence of grade ≥ 3 treatment-emergent adverse events was 22%. The decrease in ER value in sensitive patients was larger than that in non-sensitive patients, as was Ki-67 (p < 0.05). The maximum standardized uptake value, mean standardized uptake values, total lesion ER expression of 18F-FES PET-CT in sensitive patients were significantly higher than those in non-sensitive patients (p < 0.05). Moreover, these parameters were significantly correlated with Miller and Payne grade and the change in ER expression before and after treatment (p < 0.05). Thirteen differential expressed metabolites were identified, which were markedly enriched in 19 metabolic pathways. CONCLUSION This regimen demonstrated acceptable toxicity and encouraging antitumor efficacy. 18F-FES PET-CT might serve as a tool to predict the effectiveness of this therapy. Altered metabolites or metabolic pathways might be associated with treatment response.
Collapse
Affiliation(s)
- Qing Shao
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Ningning Zhang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Xianjun Pan
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Wenqi Zhou
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Yali Wang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaoliang Chen
- Department of Nuclear Medicine, Chongqing University Cancer Hospital, Chongqing, China
| | - Jing Wu
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaohua Zeng
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
3
|
Dimitrakopoulou-Strauss A, Pan L, Sachpekidis C. Non-[ 18F]FDG PET-Radiopharmaceuticals in Oncology. Pharmaceuticals (Basel) 2024; 17:1641. [PMID: 39770483 PMCID: PMC11677833 DOI: 10.3390/ph17121641] [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: 11/18/2024] [Revised: 11/26/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Molecular imaging is a growing field, driven by technological advances, such as the improvement of PET-CT scanners through the introduction of digital detectors and scanners with an extended field of view, resulting in much higher sensitivity and a variety of new specific radiopharmaceuticals that allow the visualization of specific molecular pathways and even theragnostic approaches. In oncology, the development of dedicated tracers is crucial for personalized therapeutic approaches. Novel peptides allow the visualization of many different targets, such as PD-1 and PD-L1 expression, chemokine expression, HER expression, T-cell imaging, microenvironmental imaging, such as FAP imaging, and many more. In this article, we review recent advances in the development of non-[18F]FDG PET radiopharmaceuticals and their current clinical applications in oncology, as well as some future aspects.
Collapse
Affiliation(s)
- Antonia Dimitrakopoulou-Strauss
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | | | | |
Collapse
|
4
|
Edmonds CE, O'Brien SR, McDonald ES, Mankoff DA, Pantel AR. PET Imaging of Breast Cancer: Current Applications and Future Directions. JOURNAL OF BREAST IMAGING 2024; 6:586-600. [PMID: 39401324 DOI: 10.1093/jbi/wbae053] [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: 04/23/2024] [Indexed: 11/07/2024]
Abstract
As molecular imaging use expands for patients with breast cancer, it is important for breast radiologists to have a basic understanding of molecular imaging, including PET. Although breast radiologists may not directly interpret such studies, basic knowledge of molecular imaging will enable the radiologist to better direct diagnostic workup of patients as well as discuss diagnostic imaging with the patient and other treating physicians. Several new tracers are now available to complement imaging glucose metabolism with FDG. Because it provides a noninvasive assessment of disease status across the whole body, PET offers specific advantages over tissue-based assays. Paired with targeted therapy, molecular imaging has the potential to guide personalized treatment of breast cancer, including guiding dosing during drug trials as well as predicting and assessing clinical response. This review discusses the current established applications of FDG, which remains the most widely used PET radiotracer for malignancy, including breast cancer, and highlights potential areas for expanded use based on recent research. It also summarizes research to date on the U.S. Food and Drug Administration (FDA)-approved PET tracer 16α-18F-fluoro-17β-estradiol (FES), which targets ER, including the current guidelines from the Society of Nuclear Medicine and Molecular Imaging on the appropriate use of FES-PET/CT for breast cancer as well as areas of active investigation for other potential applications. Finally, the review highlights several of the most promising novel PET tracers that are poised for clinical translation in the near future.
Collapse
Affiliation(s)
- Christine E Edmonds
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Sophia R O'Brien
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth S McDonald
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - David A Mankoff
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Austin R Pantel
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
5
|
Malik MMUD, Alqahtani MM, Hadadi I, Kanbayti I, Alawaji Z, Aloufi BA. Molecular Imaging Biomarkers for Early Cancer Detection: A Systematic Review of Emerging Technologies and Clinical Applications. Diagnostics (Basel) 2024; 14:2459. [PMID: 39518426 PMCID: PMC11545511 DOI: 10.3390/diagnostics14212459] [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: 09/09/2024] [Revised: 10/23/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Early cancer detection is crucial for improving patient outcomes. Molecular imaging biomarkers offer the potential for non-invasive, early-stage cancer diagnosis. OBJECTIVES To evaluate the effectiveness and accuracy of molecular imaging biomarkers for early cancer detection across various imaging modalities and cancer types. METHODS A comprehensive search of PubMed/MEDLINE, Embase, Web of Science, Cochrane Library, and Scopus was performed, covering the period from January 2010 to December 2023. Eligibility criteria included original research articles published in English on molecular imaging biomarkers for early cancer detection in humans. The risk of bias for included studies was evaluated using the QUADAS-2 tool. The findings were synthesized through narrative synthesis, with quantitative analysis conducted where applicable. RESULTS In total, 50 studies were included. Positron emission tomography (PET)-based biomarkers showed the highest sensitivity (mean: 89.5%, range: 82-96%) and specificity (mean: 91.2%, range: 85-100%). Novel tracers such as [68Ga]-PSMA for prostate cancer and [18F]-FES for breast cancer demonstrated promising outcomes. Optical imaging techniques showed high specificity in intraoperative settings. CONCLUSIONS Molecular imaging biomarkers show significant potential for improving early cancer detection. Integration into clinical practice could lead to earlier interventions and improved outcomes. Further research is needed to address standardization and cost-effectiveness.
Collapse
Affiliation(s)
- Maajid Mohi Ud Din Malik
- Dr. D.Y. Patil School of Allied Health Sciences, Dr. D.Y. Patil Vidyapeeth, (Deemed to be University) Sant Tukaram Nagar, Pune 411018, MH, India;
| | - Mansour M. Alqahtani
- Department of Radiological Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia;
| | - Ibrahim Hadadi
- Department of Radiological Sciences, College of Applied Medical Sciences, King Khalid University, Asir, Abha 62529, Saudi Arabia
| | - Ibrahem Kanbayti
- Radiologic Sciences Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Zeyad Alawaji
- Department of Radiologic Technology, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia;
| | - Bader A. Aloufi
- Department of Diagnostic Radiology, College of Applied Medical Sciences, Taibah University, Madinah 42353, Saudi Arabia;
| |
Collapse
|
6
|
Ryu J, Hyung J, Han S, Jeong JH, Lee SB, Yoo TKR, Kim J, Kim HJ, Chung IY, Ko BS, Lee JW, Son BH, Jeong H, Ahn JH, Jung KH, Kim SB, Moon DH. Impact of 18F-FES PET/CT on Clinical Decisions in the Management of Recurrent or Metastatic Breast Cancer. J Nucl Med 2024; 65:1689-1694. [PMID: 39362758 DOI: 10.2967/jnumed.124.267913] [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: 04/07/2024] [Accepted: 08/28/2024] [Indexed: 10/05/2024] Open
Abstract
The clinical impact of 16α-18F-fluoro-17β-estradiol (18F-FES) PET/CT on patient management has not been well investigated. The aim of this study was to assess the clinical impact of 18F-FES PET/CT on the management of patients with recurrent or metastatic breast cancer. Methods: Study subjects were identified retrospectively from a database of a prospective trial for postmarketing surveillance of 18F-FES between 2021 and 2023. Patients who were suspected or known to have recurrent or metastatic estrogen receptor-positive breast cancer based on a routine standard workup were included. Planned management before and actual management after 18F-FES PET/CT were assessed by 2 experienced medical oncologists via medical chart review. A 5-point questionnaire was provided to evaluate the value of 18F-FES PET/CT for management planning. The rate of intention-to-treat and interdisciplinary changes, and the impact of 18F-FES PET/CT according to PET/CT result or clinical indication, were examined. Results: Of the 344 included patients, 120 (35%) experienced a change in management after 18F-FES PET/CT. In 139 (40%) patients,18F-FES PET/CT supported the existing management decision without a change in management. Intention-to-treat and interdisciplinary changes accounted for 64% (77/120) and 68% (82/120) of all changes, respectively. A higher rate of change was observed when lesions were 18F-FES-negative (44% [36/81]) than 18F-FES-positive (30% [51/172]) or mixed 18F-FES-positive/negative (36% [33/91]). Regarding clinical indications, the highest rate of change was shown when evaluating the origins of metastasis of double primary cancers (64% [9/14]). Conclusion: 18F-FES PET/CT modified the management of recurrent or metastatic breast cancer, serving as an impactful imaging modality in clinical practice.
Collapse
Affiliation(s)
- Jeongryul Ryu
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jaewon Hyung
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea; and
| | - Sangwon Han
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea;
| | - Jae Ho Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea; and
| | - Sae Byul Lee
- Division of Breast Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Tae-Kyung Robyn Yoo
- Division of Breast Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jisun Kim
- Division of Breast Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hee Jeong Kim
- Division of Breast Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Il Yong Chung
- Division of Breast Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Beom Seok Ko
- Division of Breast Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jong Won Lee
- Division of Breast Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Byung Ho Son
- Division of Breast Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyehyun Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea; and
| | - Jin-Hee Ahn
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea; and
| | - Kyung Hae Jung
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea; and
| | - Sung-Bae Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea; and
| | - Dae Hyuk Moon
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
7
|
Friedel A, Prante O, Maschauer S. Radiosynthesis and Preclinical Evaluation of 18F-Labeled Estradiol Derivatives with Different Lipophilicity for PET Imaging of Breast Cancer. Cancers (Basel) 2024; 16:2639. [PMID: 39123367 PMCID: PMC11311842 DOI: 10.3390/cancers16152639] [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: 06/19/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
About 75% of breast tumors show an overexpression of the estradiol receptor (ER), making it a valuable target for tumor diagnosis and therapy. To date, 16α-[18F]fluoroestradiol (FES) is the only FDA-approved imaging probe for the positron emission tomography (PET) imaging of ER-positive (ER+) breast cancer. However, FES has the drawback of a high retention in the liver. Therefore, the aim of this study was the development and preclinical evaluation of estradiol (E2) derivatives with different lipophilicity. Three 18F-labeled prosthetic groups (two glycosyl and one PEG azide) were chosen for conjugation with ethinyl estradiol (EE) by 18F-CuAAC (Cu-catalyzed azide-alkyne cycloaddition). The cellular uptake in ER+ MCF-7 tumor cells was highest for the less hydrophilic derivative (18F-TA-Glyco-EE). In nude mice bearing different breast tumors (ER+ MCF-7 and T47D versus ER- MDA-MB-231), 18F-TA-Glyco-EE revealed a high uptake in the liver (13%ID/g, 30 min p.i.), which decreased over 90 min to 1.2%ID/g, indicating fast hepatobiliary clearance. The statistically significant difference of 18F-TA-Glyco-EE uptake in T47D compared to MDA-MB-231 tumors at 60-90 min p.i. indicated ER-specific uptake, whereas in vivo PET imaging did not provide evidence for specific uptake of 18F-TA-Glyco-EE in MCF-7 tumors, probably due to ER occupation by E2 after E2-dependent MCF-7 tumor growth in mice. However, in vitro autoradiography revealed a high specific binding of 18F-TA-Glyco-EE to ER+ tumor slices. We conclude that 18F-TA-Glyco-EE, with its increased hydrophilicity after deacetylation in the blood and thus rapid washout from non-target tissues, may be a viable alternative to FES for the PET imaging of breast cancer.
Collapse
Affiliation(s)
- Anna Friedel
- Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (A.F.); (O.P.)
| | - Olaf Prante
- Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (A.F.); (O.P.)
- FAU NeW—Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany
| | - Simone Maschauer
- Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (A.F.); (O.P.)
| |
Collapse
|
8
|
Gennari A, Brain E, De Censi A, Nanni O, Wuerstlein R, Frassoldati A, Cortes J, Rossi V, Palleschi M, Alberini JL, Matteucci F, Piccardo A, Sacchetti G, Ilhan H, D'Avanzo F, Ruffilli B, Nardin S, Monti M, Puntoni M, Fontana V, Boni L, Harbeck N. Early prediction of endocrine responsiveness in ER+/HER2-negative metastatic breast cancer (MBC): pilot study with 18F-fluoroestradiol ( 18F-FES) CT/PET. Ann Oncol 2024; 35:549-558. [PMID: 38423389 DOI: 10.1016/j.annonc.2024.02.007] [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: 06/20/2023] [Revised: 01/15/2024] [Accepted: 02/20/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND 18F-fluoroestradiol (FES) positron emission tomography (PET)/computed tomography (CT) is considered an accurate diagnostic tool to determine whole-body endocrine responsiveness. In the endocrine therapy (ET)-FES trial, we evaluated 18F-FES PET/CT as a predictive tool in estrogen receptor-positive (ER+)/human epidermal growth factor receptor 2-negative (HER2-) metastatic breast cancer (MBC). PATIENTS AND METHODS Eligible patients underwent an 18F-FES PET/CT at baseline. Patients with standardized uptake value (SUV) ≥ 2 received single-agent ET until progressive disease; patients with SUV < 2 were randomized to single-agent ET (arm A) or chemotherapy (ChT) (arm B). The primary objective was to compare the activity of first-line ET versus ChT in patients with 18F-FES SUV < 2. RESULTS Overall, 147 patients were enrolled; 117 presented with 18F-FES SUV ≥ 2 and received ET; 30 patients with SUV < 2 were randomized to ET or ChT. After a median follow-up of 62.4 months, 104 patients (73.2%) had disease progression and 53 died (37.3%). Median progression-free survival (PFS) was 12.4 months [95% confidence interval (CI) 3.1-59.6 months] in patients with SUV < 2 randomized to arm A versus 23.0 months (95% CI 7.7-30.0 months) in arm B, [hazard (HR) = 0.71, 95% CI 0.3-1.7 months]; median PFS was 18.0 months (95% CI 11.2-23.1 months) in patients with SUV ≥ 2 treated with ET. Median overall survival (OS) was 28.2 months (95% CI 14.2 months-not estimable) in patients with SUV < 2 randomized to ET (arm A) versus 52.8 months (95% CI 16.2 months-not estimable) in arm B (ChT). Median OS was not reached in patients with SUV ≥ 2. 60-month OS rate was 41.6% (95% CI 10.4% to 71.1%) in arm A, 42.0% (95% CI 14.0% to 68.2%) in arm B, and 59.6% (95% CI 48.6% to 69.0%) in patients with SUV ≥ 2. In patients with SUV ≥ 2, 60-month OS rate was 72.6% if treated with aromatase inhibitors (AIs) versus 40.6% in case of fulvestrant or tamoxifen (P < 0.005). CONCLUSIONS The ET-FES trial demonstrated that ER+/HER2- MBC patients are a heterogeneous population, with different levels of endocrine responsiveness based on 18F-FES CT/PET SUV.
Collapse
Affiliation(s)
- A Gennari
- Department of Translational Medicine, University of Piemonte Orientale, Novara; Division of Medical Oncology, Maggiore University Hospital, Novara, Italy.
| | - E Brain
- Department of Medical Oncology, Institut Curie-Hôpital René Huguenin, Saint-Cloud, France
| | - A De Censi
- Medical Oncology, E.O. Ospedali Galliera, Genova
| | - O Nanni
- Biostatistics and Clinical Trials Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - R Wuerstlein
- Department of Obstetrics and Gynecology and CCC Munich, LMU University Hospital, Munich, Germany
| | - A Frassoldati
- Clinical Oncology, S. Anna University Hospital, Ferrara, Italy
| | - J Cortes
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quironsalud Group, Barcelona; Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid, Spain
| | - V Rossi
- Division of Medical Oncology, Maggiore University Hospital, Novara, Italy
| | - M Palleschi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - J L Alberini
- Nuclear Medicine Department Centre Georges-Francois Leclerc, Dijon Cedex, France
| | - F Matteucci
- Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo studio dei tumori (IRST) -"Dino Amadori", Meldola
| | - A Piccardo
- Department of Nuclear Medicine, E.O. Ospedali Galliera, Genoa
| | - G Sacchetti
- Division of Nuclear Medicine Unit, Maggiore University Hospital, Novara, Italy
| | - H Ilhan
- Department of Nuclear Medicine, LMU University Hospital, Munich, Germany
| | - F D'Avanzo
- Division of Medical Oncology, Maggiore University Hospital, Novara, Italy
| | - B Ruffilli
- Department of Translational Medicine, University of Piemonte Orientale, Novara
| | - S Nardin
- Medical Oncology Unit 1, IRCCS-Ospedale Policlinico San Martino, Genoa
| | - M Monti
- Biostatistics and Clinical Trials Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - M Puntoni
- Clinical and Epidemiological Research Unit, University Hospital of Parma, Parma
| | - V Fontana
- Department of Clinical Epidemiology, IRCSS Ospedale Policlinico San Martino, Genoa, Italy
| | - L Boni
- Department of Clinical Epidemiology, IRCSS Ospedale Policlinico San Martino, Genoa, Italy
| | - N Harbeck
- Department of Obstetrics and Gynecology and CCC Munich, LMU University Hospital, Munich, Germany
| |
Collapse
|
9
|
Keigley QJ, Fowler AM, O'Brien SR, Dehdashti F. Molecular Imaging of Steroid Receptors in Breast Cancer. Cancer J 2024; 30:142-152. [PMID: 38753748 PMCID: PMC11101139 DOI: 10.1097/ppo.0000000000000715] [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] [Indexed: 05/18/2024]
Abstract
ABSTRACT Steroid receptors regulate gene expression for many important physiologic functions and pathologic processes. Receptors for estrogen, progesterone, and androgen have been extensively studied in breast cancer, and their expression provides prognostic information as well as targets for therapy. Noninvasive imaging utilizing positron emission tomography and radiolabeled ligands targeting these receptors can provide valuable insight into predicting treatment efficacy, staging whole-body disease burden, and identifying heterogeneity in receptor expression across different metastatic sites. This review provides an overview of steroid receptor imaging with a focus on breast cancer and radioligands for estrogen, progesterone, and androgen receptors.
Collapse
Affiliation(s)
- Quinton J Keigley
- From the Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | | | - Sophia R O'Brien
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Farrokh Dehdashti
- Division of Nuclear Medicine, Edward Mallinckrodt Institute of Radiology, Alvin J. Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| |
Collapse
|
10
|
Malviya G, Lannagan TR, Johnson E, Mackintosh A, Bielik R, Peters A, Soloviev D, Brown G, Jackstadt R, Nixon C, Gilroy K, Campbell A, Sansom OJ, Lewis DY. Noninvasive Stratification of Colon Cancer by Multiplex PET Imaging. Clin Cancer Res 2024; 30:1518-1529. [PMID: 38493804 PMCID: PMC11016897 DOI: 10.1158/1078-0432.ccr-23-1063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/30/2023] [Accepted: 02/14/2024] [Indexed: 03/19/2024]
Abstract
PURPOSE The current approach for molecular subtyping of colon cancer relies on gene expression profiling, which is invasive and has limited ability to reveal dynamics and spatial heterogeneity. Molecular imaging techniques, such as PET, present a noninvasive alternative for visualizing biological information from tumors. However, the factors influencing PET imaging phenotype, the suitable PET radiotracers for differentiating tumor subtypes, and the relationship between PET phenotypes and tumor genotype or gene expression-based subtyping remain unknown. EXPERIMENTAL DESIGN In this study, we conducted 126 PET scans using four different metabolic PET tracers, [18F]fluorodeoxy-D-glucose ([18F]FDG), O-(2-[18F]fluoroethyl)-l-tyrosine ([18F]FET), 3'-deoxy-3'-[18F]fluorothymidine ([18F]FLT), and [11C]acetate ([11C]ACE), using a spectrum of five preclinical colon cancer models with varying genetics (BMT, AKPN, AK, AKPT, KPN), at three sites (subcutaneous, orthograft, autochthonous) and at two tumor stages (primary vs. metastatic). RESULTS The results demonstrate that imaging signatures are influenced by genotype, tumor environment, and stage. PET imaging signatures exhibited significant heterogeneity, with each cancer model displaying distinct radiotracer profiles. Oncogenic Kras and Apc loss showed the most distinctive imaging features, with [18F]FLT and [18F]FET being particularly effective, respectively. The tissue environment notably impacted [18F]FDG uptake, and in a metastatic model, [18F]FET demonstrated higher uptake. CONCLUSIONS By examining factors contributing to PET-imaging phenotype, this study establishes the feasibility of noninvasive molecular stratification using multiplex radiotracer PET. It lays the foundation for further exploration of PET-based subtyping in human cancer, thereby facilitating noninvasive molecular diagnosis.
Collapse
Affiliation(s)
- Gaurav Malviya
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow; Glasgow, United Kingdom
| | | | - Emma Johnson
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
| | - Agata Mackintosh
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
| | - Robert Bielik
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
| | - Adam Peters
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
| | - Dmitry Soloviev
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
| | - Gavin Brown
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
| | - Rene Jackstadt
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Cancer Progression and Metastasis Group, German Cancer Research Center (DKFZ), and DKFZ-ZMBH Alliance, Heidelberg, Germany. German Cancer Consortium (DKTK), Germany
| | - Colin Nixon
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
| | - Kathryn Gilroy
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
| | - Andrew Campbell
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
| | - Owen J. Sansom
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow; Glasgow, United Kingdom
| | - David Y. Lewis
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow; Glasgow, United Kingdom
| |
Collapse
|
11
|
Kataoka M, Iima M, Miyake KK, Honda M. Multiparametric Approach to Breast Cancer With Emphasis on Magnetic Resonance Imaging in the Era of Personalized Breast Cancer Treatment. Invest Radiol 2024; 59:26-37. [PMID: 37994113 PMCID: PMC11805492 DOI: 10.1097/rli.0000000000001044] [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: 11/24/2023]
Abstract
ABSTRACT A multiparametric approach to breast cancer imaging offers the advantage of integrating the diverse contributions of various parameters. Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) is the most important MRI sequence for breast imaging. The vascularity and permeability of lesions can be estimated through the use of semiquantitative and quantitative parameters. The increased use of ultrafast DCE-MRI has facilitated the introduction of novel kinetic parameters. In addition to DCE-MRI, diffusion-weighted imaging provides information associated with tumor cell density, with advanced diffusion-weighted imaging techniques such as intravoxel incoherent motion, diffusion kurtosis imaging, and time-dependent diffusion MRI opening up new horizons in microscale tissue evaluation. Furthermore, T2-weighted imaging plays a key role in measuring the degree of tumor aggressiveness, which may be related to the tumor microenvironment. Magnetic resonance imaging is, however, not the only imaging modality providing semiquantitative and quantitative parameters from breast tumors. Breast positron emission tomography demonstrates superior spatial resolution to whole-body positron emission tomography and allows comparable delineation of breast cancer to MRI, as well as providing metabolic information, which often precedes vascular and morphological changes occurring in response to treatment. The integration of these imaging-derived factors is accomplished through multiparametric imaging. In this article, we explore the relationship among the key imaging parameters, breast cancer diagnosis, and histological characteristics, providing a technical and theoretical background for these parameters. Furthermore, we review the recent studies on the application of multiparametric imaging to breast cancer and the significance of the key imaging parameters.
Collapse
|
12
|
You S, Xie Y, Ji M, Liu C, Zhao Y, Gong C, Hu S, Li Y, Yang Z, Wang B. ER status conversion and subsequent treatment: an assessment of negative ER expression detected by 18F-FES PET in metastatic breast cancer patients with ER-positive primary tumors. Ther Adv Med Oncol 2023; 15:17588359231216093. [PMID: 38107829 PMCID: PMC10725097 DOI: 10.1177/17588359231216093] [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: 04/21/2023] [Accepted: 10/30/2023] [Indexed: 12/19/2023] Open
Abstract
Background The 18F-fluoroestradiol positron emission tomography/computed tomography (18F-FES PET/CT) technique provides a convenient method to evaluate the overall estrogen receptor (ER) expression in metastatic breast cancer (MBC) patients. There are long debates on the characteristics and treatment strategy of patients with positive primary ER lesions but negative ER expression in metastatic disease. 18F-FES PET offers an opportunity to answer this question. Objectives This study aimed to characterize the primary ER-positive patients with advanced-stage FES negativity and investigate the real-world treatment decisions made by physicians subsequently, and compare the efficacy between different regimens. Design This observational cohort study was conducted at Fudan University Shanghai Cancer Center, enrolling breast cancer patients with ER-positive primary tumors who showed advanced-stage FES negativity. Methods Descriptive statistics were used in clinicopathologic characteristics and compared with a chi-square test or t-test. In addition, progression-free survival (PFS) was estimated by the Kaplan-Meier method and compared by log-rank test. Results 16.6% (52/314) of patients with an ER-positive primary tumor had negative ER expression assessed by 18F-FES for MBC prior to receiving first-line systemic therapy, among whom adjuvant endocrine therapy was prevalently utilized (86.5%, 45/52). The rate of FES negativity in the advanced stage was negatively correlated with levels of ER expression of primary tumors. Chemotherapy (83.3%, 40/48) was the most common treatment strategy afterward, among which capecitabine monotherapy (62.5%, 25/40) was a dominant alternative. PFS was significantly prolonged with capecitabine alone versus other chemotherapy (median PFS: 13.14 versus 6.21 months, p = 0.029). Conclusion Negative conversion of ER in MBC detected by 18F-FES occurred frequently. Patients with lower ER expression in the primary lesion were more likely to have negative ER expression in the metastasis. In real-world clinical practice, most physicians primarily opted for chemotherapy, with capecitabine monotherapy being a commonly selected regimen. Trial registration ClinicalTrials.gov identifier: NCT05797987.
Collapse
Affiliation(s)
- Shuhui You
- Department of Breast and Urological Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yizhao Xie
- Department of Breast and Urological Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Medical Oncology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Mengjing Ji
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
- Center for Biomedical Imaging, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
| | - Cheng Liu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
- Center for Biomedical Imaging, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
| | - Yannan Zhao
- Department of Breast and Urological Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chengcheng Gong
- Department of Breast and Urological Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shihui Hu
- Department of Breast and Urological Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yumeng Li
- Department of Breast and Urological Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhongyi Yang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No. 270, Dongan Road, Shanghai 200032, China
- Center for Biomedical Imaging, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
| | - Biyun Wang
- Department of Breast and Urological Medical Oncology, Fudan University Shanghai Cancer Center, 130 Dongan Road, Xuhui District, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Liu C, Ma G, Zhang J, Cheng J, Yang Z, Song S. 18F-FES and 18F-FDG PET/CT imaging as a predictive biomarkers for metastatic breast cancer patients undergoing cyclin-dependent 4/6 kinase inhibitors with endocrine treatment. Ann Nucl Med 2023; 37:675-684. [PMID: 37787851 DOI: 10.1007/s12149-023-01871-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/19/2023] [Indexed: 10/04/2023]
Abstract
OBJECTIVE The aim of this study was to investigate the potential value of dual tracers 18F-FDG and 18F-FES PET/CT in predicting response to Cyclin-Dependent 4/6 Kinase (CDK4/6) inhibitors combined with endocrine therapy for metastatic estrogen receptor (ER)-positive breast cancer patients. METHODS This retrospective study enrolled 38 ER-positive metastatic breast cancer patients from our center who underwent both 18F-FDG and 18F-FES PET/CT scans within 1 month before CDK4/6 inhibitors combined with endocrine therapy. The extracted parameters comprised the maximum standardized uptake value (SUVmax) for both FDG and FES PET, as well as the ratio between FES and FDG SUVmax. Each parameter was dichotomized based on its median threshold. The primary endpoint was progression-free survival (PFS), which was estimated using the Kaplan-Meier method and compared by the log-rank test. RESULTS After a median follow-up of 15.6 months, progressive disease was observed in 23 out of 38 patients, and the median PFS for the whole cohort was 21.0 months [95% confidence interval (CI) 12.7-29.3]. FES and FDG PET identified 6 patients (15.8%) with FES-negative lesions, suggesting ER heterogeneity in metastatic lesions. The median PFS of these patients was only 5.3 months (95% CI 1.7-8.9), which was substantially shorter than that of patients with 100% FES-positive lesions (median PFS 22.9 months, 95% CI 17.1-28.7, P < 0.001). Patients with 100% FES-positive lesions who had high FES/FDG showed significantly shorter PFS compared to those with low FES/FDG (14.9 vs. 30.5 months, P = 0.003). CONCLUSIONS This study shows that FDG and FES PET imaging may serve as valuable tools for patient selection in the context of CDK4/6 inhibitor therapy combined with endocrine treatment, and have the potential to function as prognostic biomarkers.
Collapse
Affiliation(s)
- Cheng Liu
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, 4365 Kangxin Road, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, 200032, China
| | - Guang Ma
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jiangang Zhang
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai, 201321, China
| | - Jingyi Cheng
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai, 201321, China
| | - Zhongyi Yang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Shaoli Song
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, 4365 Kangxin Road, Shanghai, 201321, China.
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China.
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China.
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
14
|
Mahalik A, Chaudhary B, Kumar R, Tripathi M, Bal C. 18 F-FES Uptake in Radiation Pneumonitis. Clin Nucl Med 2023; 48:e468-e469. [PMID: 37566798 DOI: 10.1097/rlu.0000000000004788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2023]
Abstract
ABSTRACT 18 F-FDG uptake in radiation pneumonitis is well documented; however, the same is less so for 18 F-floroestradiol (FES), which specifically binds to the estrogen receptors in vivo. We observed increased FES uptake in the right lung of an estrogen receptor positive breast cancer patient who had undergone right modified radical mastectomy followed by radiation therapy to chest wall. The possibility of FES uptake in radiation pneumonitis must therefore be kept in mind while interpreting FES PET.
Collapse
Affiliation(s)
- Aparna Mahalik
- From the Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | | | | | | | | |
Collapse
|
15
|
Wang X, Chen C, Yan J, Xu Y, Pan D, Wang L, Yang M. Druggability of Targets for Diagnostic Radiopharmaceuticals. ACS Pharmacol Transl Sci 2023; 6:1107-1119. [PMID: 37588760 PMCID: PMC10425999 DOI: 10.1021/acsptsci.3c00081] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Indexed: 08/18/2023]
Abstract
Targets play an indispensable and pivotal role in the development of radiopharmaceuticals. However, the initial stages of drug discovery projects are often plagued by frequent failures due to inadequate information on druggability and suboptimal target selection. In this context, we aim to present a comprehensive review of the factors that influence target druggability for diagnostic radiopharmaceuticals. Specifically, we explore the crucial determinants of target specificity, abundance, localization, and positivity rate and their respective implications. Through a detailed analysis of existing protein targets, we elucidate the significance of each factor. By carefully considering and balancing these factors during the selection of targets, more efficacious and targeted radiopharmaceuticals are expected to be designed for the diagnosis of a wide range of diseases in the future.
Collapse
Affiliation(s)
- Xinyu Wang
- NHC
Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular
Nuclear Medicine, Jiangsu Institute of Nuclear
Medicine, Wuxi 214063, PR China
- School
of Pharmacy, Nanjing Medical University, Nanjing 211166, PR China
| | - Chongyang Chen
- NHC
Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular
Nuclear Medicine, Jiangsu Institute of Nuclear
Medicine, Wuxi 214063, PR China
| | - Junjie Yan
- NHC
Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular
Nuclear Medicine, Jiangsu Institute of Nuclear
Medicine, Wuxi 214063, PR China
- School
of Pharmacy, Nanjing Medical University, Nanjing 211166, PR China
| | - Yuping Xu
- NHC
Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular
Nuclear Medicine, Jiangsu Institute of Nuclear
Medicine, Wuxi 214063, PR China
- School
of Pharmacy, Nanjing Medical University, Nanjing 211166, PR China
| | - Donghui Pan
- NHC
Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular
Nuclear Medicine, Jiangsu Institute of Nuclear
Medicine, Wuxi 214063, PR China
| | - Lizhen Wang
- NHC
Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular
Nuclear Medicine, Jiangsu Institute of Nuclear
Medicine, Wuxi 214063, PR China
| | - Min Yang
- NHC
Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular
Nuclear Medicine, Jiangsu Institute of Nuclear
Medicine, Wuxi 214063, PR China
- School
of Pharmacy, Nanjing Medical University, Nanjing 211166, PR China
| |
Collapse
|
16
|
Huang YT, Chen TWW, Chen LY, Huang YY, Lu YS. The Application of 18F-FES PET in Clinical Cancer Care: A Systematic Review. Clin Nucl Med 2023:00003072-990000000-00634. [PMID: 37482660 DOI: 10.1097/rlu.0000000000004760] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
INTRODUCTION [18F]fluoroestradiol (FES) can be used for the noninvasive visualization and quantification of tumor estrogen receptor (ER) expression and activity and was FDA-approved as a diagnostic agent in May 2022 for detecting ER-positive lesions in patients with recurrent or metastatic breast cancer. PET imaging was also used to detect ER-positive lesions and malignancy among patients with uterine, ovarian, and other ER-positive solid tumors. We conducted a systemic review of the studies on FES PET imaging used among patients with cancer not limited to breast cancer to better understand the application of FES PET imaging. METHODS PubMed/MEDLINE and Cochrane Library databases were used to perform a comprehensive and systematic search and were updated until August 15, 2022. Two authors independently reviewed the titles and abstracts of the retrieved articles by using the search algorithm and selected the articles based on the inclusion and exclusion criteria. All statistical analyses were conducted using R statistical software. RESULTS Forty-three studies with 2352 patients were included in the qualitative synthesis, and 23 studies with 1388 patients were included in the quantitative analysis, which estimated the FES-positive detection rate. Thirty-two studies (77%) included breast cancer patients in 43 included studies. The FES SUVmean was higher in patients with endometrial cancer (3.4-5.3) than in those with breast cancer (2.05) and uterine sarcoma (1.1-2.6). The pooled detection rates of FES PET imaging were 0.80 for breast and 0.84 for ovarian cancer patients, both similar to that of 18F-FDG. The FES uptake threshold of 1.1 to 1.82 could detect 11.1% to 45% ER heterogeneity, but the threshold of FES uptake did not have consistent predictive ability for prognosis among patients with breast cancer, unlike uterine cancer. However, FES uptake can effectively predict and monitor treatment response, especially endocrine therapy such as estradiol, ER-blocking agents (fulvestrant and tamifoxen), and aromatase inhibitors (such as letrozole and Z-endoxifen). CONCLUSIONS [18F]fluoroestradiol PET is not only a convenient and accurate diagnostic imaging tool for detecting ER-expressing lesions in patients with breast and ovarian cancer but also among patients with uterine cancer. [18F]fluoroestradiol PET is a noninvasive predictive and monitoring tool for treatment response and prognosis.
Collapse
Affiliation(s)
| | | | - Li-Yu Chen
- From the Primo Biotechnology Co, Ltd, Taipei
| | | | | |
Collapse
|
17
|
Lin M, Jin Y, Lv H, Hu X, Zhang J. Incidence and prognostic significance of receptor discordance between primary breast cancer and paired bone metastases. Int J Cancer 2023; 152:1476-1489. [PMID: 36408915 DOI: 10.1002/ijc.34365] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 09/27/2022] [Accepted: 11/08/2022] [Indexed: 11/22/2022]
Abstract
ER, PgR and HER-2 status are the cornerstones of choosing systemic therapy for breast cancer, but can change during the disease course. Guidelines recommended the biopsy of the metastatic tumor to reassess receptor status. Bone is the most frequent metastatic site of breast cancer but remained technically difficult to biopsy. Our study aimed to evaluate the incidence and prognostic significance of receptor discordance between primary breast cancer and paired bone metastases. One hundred and fifty-five breast cancer patients were diagnosed with pathology-confirmed bone metastasis at Fudan University Shanghai Cancer Center. Ninety-three patients with receptor status available on both primary tumor and bone metastases were included in our study. ER, PgR and HER-2 status converted from positive to negative in 10.8% (10/93), 28.0% (26/93) and 8.6% (8/93) of the patients, while ER, PgR and HER-2 status converted from negative to positive in 3.2% (3/93), 4.3% (4/93) and 1.1% (1/93) of the patients, respectively. 40.4% (17/42) of the HER2-0 tumors converted to HER2-low, which enabled them to receive the treatment of new antibody-drug conjugates (ADCs). Prior endocrine and anti-HER2 therapy were the independent risk factors for receptor conversion. Loss of HR expression in bone metastases was significantly associated with worse first-line PFS (adjusted hazard ratio = 3.271, P-value = .039) and OS (adjusted hazard ratio = 6.09, P-value = .011). In conclusion, our study confirmed that patients may experience receptor conversion between primary breast cancer and bone metastases, possibly influenced by prior treatments, which significantly influenced prognosis. The rebiopsy of bone metastases in patients with primary HER2-0 tumors may benefit from the new ADC drugs.
Collapse
Affiliation(s)
- Mingxi Lin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yizi Jin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hong Lv
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xichun Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Ulaner GA, Mankoff DA, Clark AS, Fowler AM, Linden HM, Peterson LM, Dehdashti F, Kurland BF, Mortimer J, Mouabbi J, Moon DH, de Vries EGE. Summary: Appropriate Use Criteria for Estrogen Receptor-Targeted PET Imaging with 16α- 18F-Fluoro-17β-Fluoroestradiol. J Nucl Med 2023; 64:351-354. [PMID: 36863779 DOI: 10.2967/jnumed.123.265420] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 01/06/2023] [Indexed: 03/04/2023] Open
Abstract
PET imaging with 16α-18F-fluoro-17β-fluoroestradiol (18F-FES), a radiolabeled form of estradiol, allows whole-body, noninvasive evaluation of estrogen receptor (ER). 18F-FES is approved by the U.S. Food and Drug Administration as a diagnostic agent "for the detection of ER-positive lesions as an adjunct to biopsy in patients with recurrent or metastatic breast cancer." The Society of Nuclear Medicine and Molecular Imaging (SNMMI) convened an expert work group to comprehensively review the published literature for 18F-FES PET in patients with ER-positive breast cancer and to establish appropriate use criteria (AUC). The findings and discussions of the SNMMI 18F-FES work group, including example clinical scenarios, were published in full in 2022 and are available at https://www.snmmi.org/auc Of the clinical scenarios evaluated, the work group concluded that the most appropriate uses of 18F-FES PET are to assess ER functionality when endocrine therapy is considered either at initial diagnosis of metastatic breast cancer or after progression of disease on endocrine therapy, the ER status of lesions that are difficult or dangerous to biopsy, and the ER status of lesions when other tests are inconclusive. These AUC are intended to enable appropriate clinical use of 18F-FES PET, more efficient approval of FES use by payers, and promotion of investigation into areas requiring further research. This summary includes the rationale, methodology, and main findings of the work group and refers the reader to the complete AUC document.
Collapse
Affiliation(s)
- Gary A Ulaner
- Molecular Imaging and Therapy, Hoag Family Cancer Institute, Newport Beach, California;
| | - David A Mankoff
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amy S Clark
- Department of Medical Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amy M Fowler
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Hannah M Linden
- Department of Medical Oncology, University of Washington, Seattle, Washington
| | - Lanell M Peterson
- Department of Nuclear Medicine, University of Washington, Seattle, Washington
| | - Farrokh Dehdashti
- Department of Radiology, Washington University of St. Louis, St. Louis, Missouri
| | | | - Joanne Mortimer
- Department of Medical Oncology, City of Hope, Duarte, California
| | - Jason Mouabbi
- Department of Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Dae Hyuk Moon
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; and
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
19
|
O'Brien SR, Edmonds CE, Lanzo SM, Weeks JK, Mankoff DA, Pantel AR. 18F-Fluoroestradiol: Current Applications and Future Directions. Radiographics 2023; 43:e220143. [PMID: 36821506 DOI: 10.1148/rg.220143] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
In the United States, breast cancer is the second leading cause of cancer death in all women and the leading cause of cancer death in Black women. The breast cancer receptor profile, assessed with immunohistochemical staining of tissue samples, allows prediction of outcomes and direction of patient treatment. Approximately 80% of newly diagnosed breast cancers are hormone receptor (HR) positive, which is defined as estrogen receptor (ER) and/or progesterone receptor (PR) positive. Patients with ER-positive disease can be treated with therapies targeting the ER; however, the assessment of ER expression with immunohistochemical staining of biopsy specimens has several limitations including sampling error, false-negative results, challenging or inaccessible biopsy sites, and the inability to synchronously and serially assess all metastatic sites to identify spatial and/or temporal ER heterogeneity. In May 2020, after decades of research, the U.S. Food and Drug Administration approved the PET radiotracer fluorine 18 (18F) fluoroestradiol (FES) for clinical use in patients with ER-positive recurrent or metastatic breast cancer as an adjunct to biopsy. FES binds to the ER in the nucleus of ER-expressing cells, enabling whole-body in vivo assessment of ER expression. This article is focused on the approved uses of FES in the United States, including identification of a target lesion for confirmatory biopsy, in vivo assessment of biopsy-proven ER-positive disease, and evaluation of spatial and temporal ER heterogeneity. FES is an example of precision medicine that has been leveraged to optimize the care of patients with breast cancer. © RSNA, 2023 See the invited commentary by Fowler in this issue. Quiz questions for this article are available through the Online Learning Center.
Collapse
Affiliation(s)
- Sophia R O'Brien
- From the Department of Radiology, Division of Breast Imaging (S.R.O., C.E.E.) and Division of Nuclear Medicine Imaging and Therapy (S.R.O., S.M.L., J.K.W., D.A.M., A.R.P.), Hospital of the University of Pennsylvania, 3400 Spruce St, 1 Silverstein-Radiology Administration, Philadelphia, PA 19104
| | - Christine E Edmonds
- From the Department of Radiology, Division of Breast Imaging (S.R.O., C.E.E.) and Division of Nuclear Medicine Imaging and Therapy (S.R.O., S.M.L., J.K.W., D.A.M., A.R.P.), Hospital of the University of Pennsylvania, 3400 Spruce St, 1 Silverstein-Radiology Administration, Philadelphia, PA 19104
| | - Shannon M Lanzo
- From the Department of Radiology, Division of Breast Imaging (S.R.O., C.E.E.) and Division of Nuclear Medicine Imaging and Therapy (S.R.O., S.M.L., J.K.W., D.A.M., A.R.P.), Hospital of the University of Pennsylvania, 3400 Spruce St, 1 Silverstein-Radiology Administration, Philadelphia, PA 19104
| | - Joanna K Weeks
- From the Department of Radiology, Division of Breast Imaging (S.R.O., C.E.E.) and Division of Nuclear Medicine Imaging and Therapy (S.R.O., S.M.L., J.K.W., D.A.M., A.R.P.), Hospital of the University of Pennsylvania, 3400 Spruce St, 1 Silverstein-Radiology Administration, Philadelphia, PA 19104
| | - David A Mankoff
- From the Department of Radiology, Division of Breast Imaging (S.R.O., C.E.E.) and Division of Nuclear Medicine Imaging and Therapy (S.R.O., S.M.L., J.K.W., D.A.M., A.R.P.), Hospital of the University of Pennsylvania, 3400 Spruce St, 1 Silverstein-Radiology Administration, Philadelphia, PA 19104
| | - Austin R Pantel
- From the Department of Radiology, Division of Breast Imaging (S.R.O., C.E.E.) and Division of Nuclear Medicine Imaging and Therapy (S.R.O., S.M.L., J.K.W., D.A.M., A.R.P.), Hospital of the University of Pennsylvania, 3400 Spruce St, 1 Silverstein-Radiology Administration, Philadelphia, PA 19104
| |
Collapse
|
20
|
Pan B, Hao Z, Xu Y, Wang Z, Yao R, Wang X, Ren C, Zhou Y, Sun Q, Huo L. Case report: 18F-FES PET/CT predicted treatment responses of second-line and third-line CDK4/6 inhibitors after disease progression on first-line CDK4/6 inhibitor in a HR+/HER2- metastatic breast cancer patient. Front Oncol 2022; 12:1095779. [PMID: 36620595 PMCID: PMC9816999 DOI: 10.3389/fonc.2022.1095779] [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: 11/11/2022] [Accepted: 12/05/2022] [Indexed: 12/25/2022] Open
Abstract
Background Cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) has become the commonest first-line treatment of hormonal receptor positive and human epidermal growth factor receptor 2 negative (HR+/HER2-) metastatic breast cancer (MBC). However, therapy is quite individualized after progression of disease (PD) when CDK4/6i fails. Estrogen receptor (ER) status of metastatic lesions of bone, lung or liver might be different from the primary tumor and biopsy of metastatic lesions was invasive and not always available. Prediction of treatment response after PD of CDK4/6i remains unsolved. 18F-fluoroestradiol (FES) PET/CT could non-invasively reveal ER expression both in primary and metastatic breast cancer and recognize heterogeneity of ER status. Case presentation A 70-year-old woman with Parkinson's disease, osteoporosis and cardiovascular co-morbidity was diagnosed with HR+/HER2- breast cancer (pT2N2M0, stage IIIa). Three years later, she developed metastases in right lung and pleura with pleural effusion and received palbociclib + letrozole. After 8 months the disease progressed, and 18F-FES PET/CT revealed multiple ER-positive pleural lesions and ER-negative pulmonary nodules after PD and the progression-free survival (PFS) of first-line CDK4/6i was 8 months. Since most of the metastatic lesions were ER-positive, abemaciclib + fulvestrant were chosen as the second-line CDK4/6i treatment and the PFS was 15 months. Another 18F-FES PET/CT showed a new ER-positive pleural mass with multiple ER-negative pulmonary nodules. Since 18F-FES PET/CT revealed that the dominant lesions were still ER-positive, dalpiciclib + exemestane + fulvestrant were prescribed as the third-line CDK4/6i treatment. Currently the patient's disease had been stable for 2 months. Conclusion This case demonstrated that 18F-FES PET/CT could show ER heterogeneity non-invasively and reveal the treatment responses a predictive imaging tool of serial second- and third-line of CDK4/6i treatments when first-line CDK4/6i failed in HR+/HER2- MBC. So long as the dominant or newly-developed metastatic lesion was ER-positive on 18F-FES PET after first-line CDK4/6i, the patient might show certain therapeutic response towards endocrine-based treatment including second- and third-line of CDK4/6i, and thus increased the time to chemotherapy (TTC).
Collapse
Affiliation(s)
- Bo Pan
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhixin Hao
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, China
| | - Ying Xu
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhe Wang
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ru Yao
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuefei Wang
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Ren
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, China
| | - Yidong Zhou
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiang Sun
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China,*Correspondence: Qiang Sun, ; Li Huo,
| | - Li Huo
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, China,*Correspondence: Qiang Sun, ; Li Huo,
| |
Collapse
|
21
|
Kol A, Fan X, Wazynska MA, van Duijnhoven SM, Giesen D, Plat A, Van Eenennaam H, Elsinga PH, Nijman HW, de Bruyn M. Development of 89Zr-anti-CD103 PET imaging for non-invasive assessment of cancer reactive T cell infiltration. J Immunother Cancer 2022; 10:jitc-2022-004877. [PMID: 36600560 PMCID: PMC9723959 DOI: 10.1136/jitc-2022-004877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2022] [Indexed: 12/12/2022] Open
Abstract
PURPOSE CD103, an integrin specifically expressed on the surface of cancer-reactive T cells, is significantly increased during successful immunotherapy across human malignancies. In this study, we describe the generation and zirconium-89 (89Zr) radiolabeling of monoclonal antibody (mAb) clones that specifically recognize human CD103 for non-invasive immune positron-emission tomography (PET) imaging of T cell infiltration as potential biomarker for effective anticancer immune responses. EXPERIMENTAL DESIGN First, to determine the feasibility of anti-CD103 immuno-PET to visualize CD103-positive cells at physiologically and clinically relevant target densities, we developed an 89Zr-anti-murine CD103 PET tracer. Healthy, non-tumor bearing C57BL/6 mice underwent serial PET imaging after intravenous injection, followed by ex vivo biodistribution. Tracer specificity and macroscopic tissue distribution were studied using autoradiography combined with CD103 immunohistochemistry. Next, we generated and screened six unique mAbs that specifically target human CD103 positive cells. Optimal candidates were selected for 89Zr-anti-human CD103 PET development. Nude mice (BALB/cOlaHsd-Foxn1nu) with established CD103 expressing Chinese hamster ovary (CHO) or CHO wild-type xenografts were injected with 89Zr-anti-human CD103 mAbs and underwent serial PET imaging, followed by ex vivo biodistribution. RESULTS 89Zr-anti-murine CD103 PET imaging identified CD103-positive tissues at clinically relevant target densities. For human anti-human CD103 PET development two clones were selected based on strong binding to the CD103+ CD8+ T cell subpopulation in ovarian cancer tumor digests, non-overlapping binding epitopes and differential CD103 blocking properties. In vivo, both 89Zr-anti-human CD103 tracers showed high target-to-background ratios, high target site selectivity and a high sensitivity in human CD103 positive xenografts. CONCLUSION CD103 immuno-PET tracers visualize CD103 T cells at relevant densities and are suitable for future non-invasive assessment of cancer reactive T cell infiltration.
Collapse
Affiliation(s)
- Arjan Kol
- Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Xiaoyu Fan
- Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marta A. Wazynska
- Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Danique Giesen
- Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Annechien Plat
- Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Philip H. Elsinga
- Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hans W. Nijman
- Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marco de Bruyn
- Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
22
|
Goodman K, Abel MK, Lawhn-Heath C, Molina-Vega J, Jones EF, Mukhtar RA. Molecular Imaging for Estrogen Receptor-Positive Breast Cancer. Surg Oncol Clin N Am 2022; 31:569-579. [DOI: 10.1016/j.soc.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
23
|
Van Baelen K, Geukens T, Maetens M, Tjan-Heijnen V, Lord CJ, Linn S, Bidard FC, Richard F, Yang WW, Steele RE, Pettitt SJ, Van Ongeval C, De Schepper M, Isnaldi E, Nevelsteen I, Smeets A, Punie K, Voorwerk L, Wildiers H, Floris G, Vincent-Salomon A, Derksen PWB, Neven P, Senkus E, Sawyer E, Kok M, Desmedt C. Current and future diagnostic and treatment strategies for patients with invasive lobular breast cancer. Ann Oncol 2022; 33:769-785. [PMID: 35605746 DOI: 10.1016/j.annonc.2022.05.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/06/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Invasive lobular breast cancer (ILC) is the second most common type of breast cancer after invasive breast cancer of no special type (NST), representing up to 15% of all breast cancers. DESIGN Latest data on ILC are presented, focusing on diagnosis, molecular make-up according to the European Society for Medical Oncology Scale for Clinical Actionability of molecular Targets (ESCAT) guidelines, treatment in the early and metastatic setting and ILC-focused clinical trials. RESULTS At the imaging level, magnetic resonance imaging-based and novel positron emission tomography/computed tomography-based techniques can overcome the limitations of currently used imaging techniques for diagnosing ILC. At the pathology level, E-cadherin immunohistochemistry could help improving inter-pathologist agreement. The majority of patients with ILC do not seem to benefit as much from (neo-)adjuvant chemotherapy as patients with NST, although chemotherapy might be required in a subset of high-risk patients. No differences in treatment efficacy are seen for anti-human epidermal growth factor receptor 2 (HER2) therapies in the adjuvant setting and cyclin-dependent kinases 4 and 6 inhibitors in the metastatic setting. The clinical utility of the commercially available prognostic gene expression-based tests is unclear for patients with ILC. Several ESCAT alterations differ in frequency between ILC and NST. Germline BRCA1 and PALB2 alterations are less frequent in patients with ILC, while germline CDH1 (gene coding for E-cadherin) alterations are more frequent in patients with ILC. Somatic HER2 mutations are more frequent in ILC, especially in metastases (15% ILC versus 5% NST). A high tumour mutational burden, relevant for immune checkpoint inhibition, is more frequent in ILC metastases (16%) than in NST metastases (5%). Tumours with somatic inactivating CDH1 mutations may be vulnerable for treatment with ROS1 inhibitors, a concept currently investigated in early and metastatic ILC. CONCLUSION ILC is a unique malignancy based on its pathological and biological features leading to differences in diagnosis as well as in treatment response, resistance and targets as compared to NST.
Collapse
Affiliation(s)
- K Van Baelen
- Laboratory for Translational Breast Cancer Research (LTBCR), Department of Oncology, KU Leuven, Leuven, Belgium; Departments of Gynaecology and Obstetrics, UZ Leuven, Leuven, Belgium
| | - T Geukens
- Laboratory for Translational Breast Cancer Research (LTBCR), Department of Oncology, KU Leuven, Leuven, Belgium; General Medical Oncology, UZ Leuven, Leuven, Belgium
| | - M Maetens
- Laboratory for Translational Breast Cancer Research (LTBCR), Department of Oncology, KU Leuven, Leuven, Belgium
| | - V Tjan-Heijnen
- Medical Oncology Department, Maastricht University Medical Center (MUMC), School of GROW, Maastricht, The Netherlands
| | - C J Lord
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - S Linn
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands; Departments of Medical Oncology, Amsterdam, The Netherlands; Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - F-C Bidard
- Department of Medical Oncology, Institut Curie, UVSQ/Paris-Saclav University, Paris, France
| | - F Richard
- Laboratory for Translational Breast Cancer Research (LTBCR), Department of Oncology, KU Leuven, Leuven, Belgium
| | - W W Yang
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - R E Steele
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - S J Pettitt
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - C Van Ongeval
- Departments of Radiology, UZ Leuven, Leuven, Belgium
| | - M De Schepper
- Laboratory for Translational Breast Cancer Research (LTBCR), Department of Oncology, KU Leuven, Leuven, Belgium; Pathology, UZ Leuven, Leuven, Belgium
| | - E Isnaldi
- Laboratory for Translational Breast Cancer Research (LTBCR), Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - A Smeets
- Surgical Oncology, UZ Leuven, Leuven, Belgium
| | - K Punie
- General Medical Oncology, UZ Leuven, Leuven, Belgium
| | - L Voorwerk
- Departments of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Tumour Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - H Wildiers
- General Medical Oncology, UZ Leuven, Leuven, Belgium
| | - G Floris
- Pathology, UZ Leuven, Leuven, Belgium
| | | | - P W B Derksen
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - P Neven
- Departments of Gynaecology and Obstetrics, UZ Leuven, Leuven, Belgium
| | - E Senkus
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - E Sawyer
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, Guy's Cancer Centre, King's College London, London, UK
| | - M Kok
- Departments of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Tumour Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - C Desmedt
- Laboratory for Translational Breast Cancer Research (LTBCR), Department of Oncology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
24
|
Yadav D, Kumar R, Phulia A, Basu S, Alavi A. Molecular Imaging Assessment of Hormonally Sensitive Breast Cancer: An Appraisal of 2-[18F]-Fluoro-2-Deoxy-Glucose and Newer Non-2-[18F]-Fluoro-2-Deoxy-Glucose PET Tracers. PET Clin 2022; 17:399-413. [PMID: 35717099 DOI: 10.1016/j.cpet.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Hormone-sensitive breast cancer, which demonstrates hormone receptor positivity, accounts for approximately 75% of newly diagnosed breast cancer. 2-[18F]-Fluoro-2-deoxy-glucose is the nonspecific radiotracer of glucose metabolism as opposed to specific receptor based tracers like 16α-[18F]-fluoro-17β-estradiol and [18F]-fluoro-furanyl-norprogesterone, which provide essential information about receptor status in the management of hormonally active malignancies. The complementary information provided by (a) 2-[18F]-fluoro-2-deoxy-glucose imaging for staging and prognostication along with (b) analyzing the hormonal receptor status with receptor-based PET imaging in breast cancer can optimize tumor characterization and influence patient management.
Collapse
Affiliation(s)
- Divya Yadav
- MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Rakesh Kumar
- Division of Diagnostic Nuclear Medicine, Department of Nuclear Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India.
| | - Ankita Phulia
- Maulana Azad Medical College, New Delhi, 110002, India
| | - Sandip Basu
- Radiation Medicine Centre (B.A.R.C), Tata Memorial Centre Annexe, Parel, Mumbai; Homi Bhabha National Institute, Mumbai, India
| | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
25
|
Diagnostic and Prognostic Role of 18F-Fluoroestradiol PET in Metastatic Breast Cancer: The Second Youth of an Older Theranostic Concept. J Clin Med 2022; 11:jcm11133589. [PMID: 35806878 PMCID: PMC9267420 DOI: 10.3390/jcm11133589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
Since the discovery of the role of female hormones in breast cancer (BC) pathophysiology, in vivo detection of oestrogen receptor (ER) distribution has been one of the major goals of nuclear medicine and molecular imaging [...]
Collapse
|
26
|
Edmonds CE, O'Brien SR, Mankoff DA, Pantel AR. Novel applications of molecular imaging to guide breast cancer therapy. Cancer Imaging 2022; 22:31. [PMID: 35729608 PMCID: PMC9210593 DOI: 10.1186/s40644-022-00468-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/30/2022] [Indexed: 11/10/2022] Open
Abstract
The goals of precision oncology are to provide targeted drug therapy based on each individual’s specific tumor biology, and to enable the prediction and early assessment of treatment response to allow treatment modification when necessary. Thus, precision oncology aims to maximize treatment success while minimizing the side effects of inadequate or suboptimal therapies. Molecular imaging, through noninvasive assessment of clinically relevant tumor biomarkers across the entire disease burden, has the potential to revolutionize clinical oncology, including breast oncology. In this article, we review breast cancer positron emission tomography (PET) imaging biomarkers for providing early response assessment and predicting treatment outcomes. For 2-18fluoro-2-deoxy-D-glucose (FDG), a marker of cellular glucose metabolism that is well established for staging multiple types of malignancies including breast cancer, we highlight novel applications for early response assessment. We then review current and future applications of novel PET biomarkers for imaging the steroid receptors, including the estrogen and progesterone receptors, the HER2 receptor, cellular proliferation, and amino acid metabolism.
Collapse
Affiliation(s)
- Christine E Edmonds
- Department of Radiology, Hospital of the University if Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA.
| | - Sophia R O'Brien
- Department of Radiology, Hospital of the University if Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - David A Mankoff
- Department of Radiology, Hospital of the University if Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Austin R Pantel
- Department of Radiology, Hospital of the University if Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| |
Collapse
|
27
|
Covington MF, Koppula BR, Fine GC, Salem AE, Wiggins RH, Hoffman JM, Morton KA. PET-CT in Clinical Adult Oncology: II. Primary Thoracic and Breast Malignancies. Cancers (Basel) 2022; 14:cancers14112689. [PMID: 35681669 PMCID: PMC9179296 DOI: 10.3390/cancers14112689] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/20/2022] [Accepted: 05/26/2022] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Positron emission tomography (PET), typically combined with computed tomography (CT), has become a critical advanced imaging technique in oncology. With PET-CT, a radioactive molecule (radiotracer) is injected in the bloodstream and localizes to sites of tumor because of specific cellular features of the tumor that accumulate the targeting radiotracer. The CT scan, performed at the same time, provides information to facilitate assessment of the amount of radioactivity from deep or dense structures, and to provide detailed anatomic information. PET-CT has a variety of applications in oncology, including staging, therapeutic response assessment, restaging, and surveillance. This series of six review articles provides an overview of the value, applications, and imaging and interpretive strategies of PET-CT in the more common adult malignancies. The second article in this series addresses the use of PET-CT in breast cancer and other primary thoracic malignancies. Abstract Positron emission tomography combined with x-ray computed tomography (PET-CT) is an advanced imaging modality with oncologic applications that include staging, therapy assessment, restaging, and surveillance. This six-part series of review articles provides practical information to providers and imaging professionals regarding the best use of PET-CT for the more common adult malignancies. The second article of this series addresses primary thoracic malignancy and breast cancer. For primary thoracic malignancy, the focus will be on lung cancer, malignant pleural mesothelioma, thymoma, and thymic carcinoma, with an emphasis on the use of FDG PET-CT. For breast cancer, the various histologic subtypes will be addressed, and will include 18F fluorodeoxyglucose (FDG), recently Food and Drug Administration (FDA)-approved 18F-fluoroestradiol (FES), and 18F sodium fluoride (NaF). The pitfalls and nuances of PET-CT in breast and primary thoracic malignancies and the imaging features that distinguish between subcategories of these tumors are addressed. This review will serve as a resource for the appropriate roles and limitations of PET-CT in the clinical management of patients with breast and primary thoracic malignancies for healthcare professionals caring for adult patients with these cancers. It also serves as a practical guide for imaging providers, including radiologists, nuclear medicine physicians, and their trainees.
Collapse
Affiliation(s)
- Matthew F. Covington
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (M.F.C.); (B.R.K.); (G.C.F.); (A.E.S.); (R.H.W.); (J.M.H.)
| | - Bhasker R. Koppula
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (M.F.C.); (B.R.K.); (G.C.F.); (A.E.S.); (R.H.W.); (J.M.H.)
| | - Gabriel C. Fine
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (M.F.C.); (B.R.K.); (G.C.F.); (A.E.S.); (R.H.W.); (J.M.H.)
| | - Ahmed Ebada Salem
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (M.F.C.); (B.R.K.); (G.C.F.); (A.E.S.); (R.H.W.); (J.M.H.)
- Department of Radiodiagnosis and Intervention, Faculty of Medicine, Alexandria University, Alexandria 21526, Egypt
| | - Richard H. Wiggins
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (M.F.C.); (B.R.K.); (G.C.F.); (A.E.S.); (R.H.W.); (J.M.H.)
| | - John M. Hoffman
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (M.F.C.); (B.R.K.); (G.C.F.); (A.E.S.); (R.H.W.); (J.M.H.)
| | - Kathryn A. Morton
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (M.F.C.); (B.R.K.); (G.C.F.); (A.E.S.); (R.H.W.); (J.M.H.)
- Intermountain Healthcare Hospitals, Summit Physician Specialists, Murray, UT 84123, USA
- Correspondence: ; Tel.: +1-801-581-7553
| |
Collapse
|
28
|
van Geel JJL, Boers J, Elias SG, Glaudemans AWJM, de Vries EFJ, Hospers GAP, van Kruchten M, Kuip EJM, Jager A, Menke-van der Houven van Oordt WC, van der Vegt B, de Vries EGE, Schröder CP. Clinical Validity of 16α-[ 18F]Fluoro-17β-Estradiol Positron Emission Tomography/Computed Tomography to Assess Estrogen Receptor Status in Newly Diagnosed Metastatic Breast Cancer. J Clin Oncol 2022; 40:3642-3652. [PMID: 35584346 DOI: 10.1200/jco.22.00400] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
PURPOSE Determining the estrogen receptor (ER) status is essential in metastatic breast cancer (MBC) management. Whole-body ER imaging with 16α-[18F]fluoro-17β-estradiol positron emission tomography ([18F]FES-PET) is increasingly used for this purpose. To establish the clinical validity of the [18F]FES-PET, we studied the diagnostic accuracy of qualitative and quantitative [18F]FES-PET assessment to predict ER expression by immunohistochemistry in a metastasis. METHODS In a prospective multicenter trial, 200 patients with newly diagnosed MBC underwent extensive workup including molecular imaging. For this subanalysis, ER expression in the biopsied metastasis was related to qualitative whole-body [18F]FES-PET evaluation and quantitative [18F]FES uptake in the corresponding metastasis. A review and meta-analysis regarding [18F]FES-PET diagnostic performance were performed. RESULTS Whole-body [18F]FES-PET assessment predicted ER expression in the biopsied metastasis with good accuracy: a sensitivity of 95% (95% CI, 89 to 97), a specificity of 80% (66 to 89), a positive predictive value (PPV) of 93% (87 to 96), and a negative predictive value (NPV) of 85% (72 to 92) in 181 of 200 evaluable patients. Quantitative [18F]FES uptake predicted ER immunohistochemistry in the corresponding metastasis with a sensitivity/specificity of 91%/69% and a PPV/NPV of 90%/71% in 156 of 200 evaluable patients. For bone metastases, PPV/NPV was 92%/81%. Meta-analysis with addition of our data has increased diagnostic performance and narrowed the 95% CIs compared with previous studies with a sensitivity/specificity of both 86% (81 to 90 and 73 to 93, respectively). CONCLUSION In this largest prospective series so far, we established the clinical validity of [18F]FES-PET to determine tumor ER status in MBC. In view of the high diagnostic accuracy of qualitatively assessed whole-body [18F]FES-PET, this noninvasive imaging modality can be considered a valid alternative to a biopsy of a metastasis to determine ER status in newly MBC (ClinicalTrials.gov identifier: NCT01957332).
Collapse
Affiliation(s)
- Jasper J L van Geel
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jorianne Boers
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Sjoerd G Elias
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Andor W J M Glaudemans
- Department of Nuclear Medicine and Molecular Imaging, 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
| | - Geke A P Hospers
- Department of Medical Oncology, 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
| | - Evelien J M Kuip
- Department of Medical Oncology, Radboud Medical Center, Nijmegen, the Netherlands
| | - Agnes Jager
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | - Bert van der Vegt
- Department of Pathology, 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, Dutch Cancer Institute, Amsterdam and University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
29
|
O’Brien SR, Edmonds CE, Katz D, Mankoff DA, Pantel AR. 18F-Fluoroestradiol (FES) PET/CT: review of current practice and future directions. Clin Transl Imaging 2022. [DOI: 10.1007/s40336-022-00494-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
30
|
Ulaner GA. 16α-18F-fluoro-17β-Fluoroestradiol (FES): Clinical Applications for Patients With Breast Cancer. Semin Nucl Med 2022; 52:574-583. [DOI: 10.1053/j.semnuclmed.2022.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 02/22/2022] [Accepted: 03/01/2022] [Indexed: 12/25/2022]
|
31
|
Kumar M, Salem K, Jeffery JJ, Fowler AM. PET Imaging of Estrogen Receptors Using 18F-Based Radioligands. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2418:129-151. [PMID: 35119664 DOI: 10.1007/978-1-0716-1920-9_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In vivo molecular imaging of estrogen receptor alpha (ER) can be performed via positron emission tomography (PET) using ER-specific radioligands, such as 16α-[18F]fluoro-17β-estradiol (18F-FES). 18F-FES is a radiopharmaceutical recently approved by the United States Food and Drug Administration for use with PET imaging to detect ER+ lesions in patients with recurrent or metastatic breast cancer as an adjunct to biopsy. 18F-FES PET imaging has been used in clinical studies and preclinical research to assess whole-body ER protein expression and ligand binding function across multiple metastatic sites, to demonstrate inter-tumoral and temporal heterogeneity of ER expression, to quantify the pharmacodynamic effects of ER antagonist treatment, and to predict endocrine therapy response. 18F-FES PET has also been studied for imaging ER in endometrial and ovarian cancer. This chapter details the experimental protocol for 18F-FES PET imaging of ER in preclinical tumor xenograft models. Consistent adherence to key methodologic details will facilitate obtaining meaningful and reproducible 18F-FES PET preclinical imaging results, which could yield additional insight for clinical trials regarding imaging biomarkers and oncologic therapy.
Collapse
Affiliation(s)
- Manoj Kumar
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA, USA
| | - Kelley Salem
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | - Amy M Fowler
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA.
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
32
|
Vahidfar N, Farzanefar S, Ahmadzadehfar H, Molloy EN, Eppard E. A Review of Nuclear Medicine Approaches in the Diagnosis and the Treatment of Gynecological Malignancies. Cancers (Basel) 2022; 14:1779. [PMID: 35406552 PMCID: PMC8997132 DOI: 10.3390/cancers14071779] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/19/2022] [Accepted: 03/26/2022] [Indexed: 12/15/2022] Open
Abstract
Nuclear medicine is defined as the diagnosis and the treatment of disease using radiolabeled compounds known as radiopharmaceuticals. Single-photon emission computed tomography/computed tomography (SPECT/CT) and positron emission tomography/computer tomography (PET/CT) based radiopharmaceuticals have proven reliable in diagnostic imaging in nuclear medicine and cancer treatment. One of the most critical cancers that also relies on an early diagnosis is gynecological cancer. Given that approximately 25% of all cancers in developing countries are a subset of gynecological cancer, investigating this cancer subtype is of significant clinical worth, particularly in light of its high rate of mortality. With accurate identification of high grade distant abdominal endometrial cancer as well as extra abdominal metastases, 18F-Fluorodeoxyglucose ([18F]FDG) PET/CT imaging is considered a valuable step forward in the investigation of gynecological cancer. Considering these factors, [18F]FDG PET/CT imaging can assist in making management of patient therapy more feasible. In this literature review, we will provide a short overview of the role of nuclear medicine in the diagnosis of obstetric and gynecological cancers.
Collapse
Affiliation(s)
- Nasim Vahidfar
- Department of Nuclear Medicine, Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran 1419733133, Iran; (N.V.); (S.F.)
| | - Saeed Farzanefar
- Department of Nuclear Medicine, Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran 1419733133, Iran; (N.V.); (S.F.)
| | | | - Eóin N. Molloy
- University Clinic for Radiology and Nuclear Medicine, Faculty of Medicine, Otto von Guericke University (OvGU), 39120 Magdeburg, Germany;
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Elisabeth Eppard
- University Clinic for Radiology and Nuclear Medicine, Faculty of Medicine, Otto von Guericke University (OvGU), 39120 Magdeburg, Germany;
| |
Collapse
|
33
|
Piccardo A, Fiz F, Treglia G, Bottoni G, Trimboli P. Head-to-Head Comparison between 18F-FES PET/CT and 18F-FDG PET/CT in Oestrogen Receptor-Positive Breast Cancer: A Systematic Review and Meta-Analysis. J Clin Med 2022; 11:jcm11071919. [PMID: 35407526 PMCID: PMC8999922 DOI: 10.3390/jcm11071919] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/18/2022] [Accepted: 03/28/2022] [Indexed: 12/26/2022] Open
Abstract
18F-FDG PET/CT is a powerful diagnostic tool in breast cancer (BC). However, it might have a reduced sensitivity in differentiated, oestrogen receptor-positive (ER+) BC. In this setting, specific molecular imaging with fluorine-oestradiol (18F-FES) PET/CT could help in overcoming these limitations; however, the literature on the diagnostic accuracy of this method is limited. We therefore planned this systematic review and meta-analysis to compare 18F-FDG and 18F-FES PET/CT in ER+ BC patients. We performed a literature search to identify all studies performing a head-to-head comparison between the two methods; we excluded review articles, preclinical studies, case reports and small case series. Finally, seven studies were identified (overall: 171 patients; range: 7–49 patients). A patients-based analysis (PBA) showed that 18F-FDG and 18F-FES PET/CT had a similar high pooled sensitivity (97% and 94%, respectively) at the lesion-based analysis (LBA), 18F-FES performed slightly better than 18F-FDG (pooled sensitivity: 95% vs. 85%, respectively). Moreover, when we considered only the studies dealing with the restaging setting (n = 3), this difference in sensitivity was even more marked (98% vs. 81%, respectively). In conclusion, both tracers feature an excellent sensitivity in ER+ BC; however, 18F-FES PET/CT could be preferred in the restaging setting.
Collapse
Affiliation(s)
- Arnoldo Piccardo
- Nuclear Medicine Department, Ente Ospedaliero “Ospedali Galliera”, 16128 Genoa, Italy; (F.F.); (G.B.)
- Correspondence: ; Tel.: +39-(0)10-5634541
| | - Francesco Fiz
- Nuclear Medicine Department, Ente Ospedaliero “Ospedali Galliera”, 16128 Genoa, Italy; (F.F.); (G.B.)
| | - Giorgio Treglia
- Faculty of Biomedical Sciences, University della Svizzera Italiana (USI), 6900 Lugano, Switzerland; (G.T.); (P.T.)
- Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
- Academic Education, Research and Innovation Area, General Directorate, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
| | - Gianluca Bottoni
- Nuclear Medicine Department, Ente Ospedaliero “Ospedali Galliera”, 16128 Genoa, Italy; (F.F.); (G.B.)
| | - Pierpaolo Trimboli
- Faculty of Biomedical Sciences, University della Svizzera Italiana (USI), 6900 Lugano, Switzerland; (G.T.); (P.T.)
- Clinic of Endocrinology and Diabetology, Lugano and Mendrisio Regional Hospital, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
| |
Collapse
|
34
|
De Leo TC, Dos Santos SN, Bernardes ES, Cummings RD, Stowell SR, Dias-Baruffi M. Molecular Imaging for In Vivo Tracking and Detection of Galectin Binding Partners. Methods Mol Biol 2022; 2442:339-352. [PMID: 35320534 DOI: 10.1007/978-1-0716-2055-7_18] [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: 06/14/2023]
Abstract
Molecular imaging (MI) is a non-invasive growing technology that allows the investigation of cellular and molecular processes in basic and clinical research and medicine. Luminescent proteins and radionuclides can be associated to target molecules providing high-definition and real-time image of whole body in few minutes or hours. Several MI studies have enabled the determination of molecular partners, in vivo tracking, and fate of compounds in different disorders. Considering that galectins are multifaceted proteins with great impact in many biological events, here we describe methods and strategies to generate labeled galectins for in vivo non-invasive imaging studies.
Collapse
Affiliation(s)
- Thais Canassa De Leo
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Sofia Nascimento Dos Santos
- Departamento de Radiofarmácia, Instituto de Pesquisas Energéticas e Nucleares, IPEN/CNEN, São Paulo, SP, Brasil
| | - Emerson Soares Bernardes
- Departamento de Radiofarmácia, Instituto de Pesquisas Energéticas e Nucleares, IPEN/CNEN, São Paulo, SP, Brasil
| | | | - Sean R Stowell
- Harvard Glycomics Center, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marcelo Dias-Baruffi
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil.
| |
Collapse
|
35
|
Oprea-Lager DE, Cysouw MC, Boellaard R, Deroose CM, de Geus-Oei LF, Lopci E, Bidaut L, Herrmann K, Fournier LS, Bäuerle T, deSouza NM, Lecouvet FE. Bone Metastases Are Measurable: The Role of Whole-Body MRI and Positron Emission Tomography. Front Oncol 2021; 11:772530. [PMID: 34869009 PMCID: PMC8640187 DOI: 10.3389/fonc.2021.772530] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/04/2021] [Indexed: 12/14/2022] Open
Abstract
Metastatic tumor deposits in bone marrow elicit differential bone responses that vary with the type of malignancy. This results in either sclerotic, lytic, or mixed bone lesions, which can change in morphology due to treatment effects and/or secondary bone remodeling. Hence, morphological imaging is regarded unsuitable for response assessment of bone metastases and in the current Response Evaluation Criteria In Solid Tumors 1.1 (RECIST1.1) guideline bone metastases are deemed unmeasurable. Nevertheless, the advent of functional and molecular imaging modalities such as whole-body magnetic resonance imaging (WB-MRI) and positron emission tomography (PET) has improved the ability for follow-up of bone metastases, regardless of their morphology. Both these modalities not only have improved sensitivity for visual detection of bone lesions, but also allow for objective measurements of bone lesion characteristics. WB-MRI provides a global assessment of skeletal metastases and for a one-step "all-organ" approach of metastatic disease. Novel MRI techniques include diffusion-weighted imaging (DWI) targeting highly cellular lesions, dynamic contrast-enhanced MRI (DCE-MRI) for quantitative assessment of bone lesion vascularization, and multiparametric MRI (mpMRI) combining anatomical and functional sequences. Recommendations for a homogenization of MRI image acquisitions and generalizable response criteria have been developed. For PET, many metabolic and molecular radiotracers are available, some targeting tumor characteristics not confined to cancer type (e.g. 18F-FDG) while other targeted radiotracers target specific molecular characteristics, such as prostate specific membrane antigen (PSMA) ligands for prostate cancer. Supporting data on quantitative PET analysis regarding repeatability, reproducibility, and harmonization of PET/CT system performance is available. Bone metastases detected on PET and MRI can be quantitatively assessed using validated methodologies, both on a whole-body and individual lesion basis. Both have the advantage of covering not only bone lesions but visceral and nodal lesions as well. Hybrid imaging, combining PET with MRI, may provide complementary parameters on the morphologic, functional, metabolic and molecular level of bone metastases in one examination. For clinical implementation of measuring bone metastases in response assessment using WB-MRI and PET, current RECIST1.1 guidelines need to be adapted. This review summarizes available data and insights into imaging of bone metastases using MRI and PET.
Collapse
Affiliation(s)
- Daniela E. Oprea-Lager
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Matthijs C.F. Cysouw
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Christophe M. Deroose
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
- Nuclear Medicine & Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
- Biomedical Photonic Imaging Group, University of Twente, Enschede, Netherlands
| | - Egesta Lopci
- Nuclear Medicine Unit, IRCCS – Humanitas Research Hospital, Milan, Italy
| | - Luc Bidaut
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- College of Science, University of Lincoln, Lincoln, United Kingdom
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen, and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Laure S. Fournier
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Paris Cardiovascular Research Center (PARCC), Institut National de la Santé et de la Recherche Médicale (INSERM), Radiology Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Hopital europeen Georges Pompidou, Université de Paris, Paris, France
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
| | - Tobias Bäuerle
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Nandita M. deSouza
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Frederic E. Lecouvet
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology, Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
36
|
Boers J, Loudini N, de Haas RJ, Willemsen ATM, van der Vegt B, de Vries EGE, Hospers GAP, Schröder CP, Glaudemans AWJM, de Vries EFJ. Analyzing the Estrogen Receptor Status of Liver Metastases with [ 18F]-FES-PET in Patients with Breast Cancer. Diagnostics (Basel) 2021; 11:diagnostics11112019. [PMID: 34829366 PMCID: PMC8617814 DOI: 10.3390/diagnostics11112019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/03/2021] [Accepted: 10/21/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Positron emission tomography (PET) with 16α-[18F]-fluoro-17β-estradiol ([18F]-FES) can visualize estrogen receptor (ER) expression, but it is challenging to determine the ER status of liver metastases, due to high physiological [18F]-FES uptake. We evaluated whether [18F]-FES-PET can be used to determine the ER status of liver metastases, using corresponding liver biopsies as the gold standard. Methods: Patients with metastatic breast cancer (n = 23) were included if they had undergone a [18F]-FES-PET, liver metastasis biopsy, CT-scan, and [18F]-FDG-PET. [18F]-FES-PET scans were assessed by visual and quantitative analysis, tracer uptake was correlated with ER expression measured by immunohistochemical staining and the effects of region-of-interest size and background correction were determined. Results: Visual analysis allowed ER assessment of liver metastases with 100% specificity and 18% sensitivity. Quantitative analysis improved the sensitivity. Reduction of the region-of-interest size did not further improve the results, but background correction improved ER assessment, resulting in 83% specificity and 77% sensitivity. Using separate thresholds for ER+ and ER− metastases, positive and negative predictive values of 100% and 75%, respectively, could be obtained, although 30% of metastases remained inconclusive. Conclusion: In the majority of liver metastases, ER status can be determined with [18F]-FES-PET if background correction and separate thresholds are applied.
Collapse
Affiliation(s)
- Jorianne Boers
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, 9713 Groningen, The Netherlands; (J.B.); (E.G.E.d.V.); (G.A.P.H.); (C.P.S.)
| | - Naila Loudini
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 Groningen, The Netherlands; (N.L.); (A.T.M.W.); (A.W.J.M.G.)
| | - Robbert J. de Haas
- Department of Radiology, University Medical Center Groningen, University of Groningen, 9713 Groningen, The Netherlands;
| | - Antoon T. M. Willemsen
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 Groningen, The Netherlands; (N.L.); (A.T.M.W.); (A.W.J.M.G.)
| | - Bert van der Vegt
- Department of Pathology, University Medical Center Groningen, University of Groningen, 9713 Groningen, The Netherlands;
| | - Elisabeth G. E. de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, 9713 Groningen, The Netherlands; (J.B.); (E.G.E.d.V.); (G.A.P.H.); (C.P.S.)
| | - Geke A. P. Hospers
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, 9713 Groningen, The Netherlands; (J.B.); (E.G.E.d.V.); (G.A.P.H.); (C.P.S.)
| | - Carolina P. Schröder
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, 9713 Groningen, The Netherlands; (J.B.); (E.G.E.d.V.); (G.A.P.H.); (C.P.S.)
| | - Andor W. J. M. Glaudemans
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 Groningen, The Netherlands; (N.L.); (A.T.M.W.); (A.W.J.M.G.)
| | - Erik F. J. de Vries
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 Groningen, The Netherlands; (N.L.); (A.T.M.W.); (A.W.J.M.G.)
- Correspondence: ; Tel.: +31-50-36-13599
| |
Collapse
|
37
|
Liu F, Dong J, Shen Y, Yun C, Wang R, Wang G, Tan J, Wang T, Yao Q, Wang B, Li L, Mi J, Zhou D, Xiong F. Comparison of PET/CT and MRI in the Diagnosis of Bone Metastasis in Prostate Cancer Patients: A Network Analysis of Diagnostic Studies. Front Oncol 2021; 11:736654. [PMID: 34671558 PMCID: PMC8522477 DOI: 10.3389/fonc.2021.736654] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/10/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Accurate diagnosis of bone metastasis status of prostate cancer (PCa) is becoming increasingly more important in guiding local and systemic treatment. Positron emission tomography/computed tomography (PET/CT) and magnetic resonance imaging (MRI) have increasingly been utilized globally to assess the bone metastases in PCa. Our meta-analysis was a high-volume series in which the utility of PET/CT with different radioligands was compared to MRI with different parameters in this setting. MATERIALS AND METHODS Three databases, including Medline, Embase, and Cochrane Library, were searched to retrieve original trials from their inception to August 31, 2019 according to the Preferred Reporting Items for Systematic Review and Meta-analysis (PRISMA) statement. The methodological quality of the included studies was assessed by two independent investigators utilizing Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2). A Bayesian network meta-analysis was performed using an arm-based model. Absolute sensitivity and specificity, relative sensitivity and specificity, diagnostic odds ratio (DOR), and superiority index, and their associated 95% confidence intervals (CI) were used to assess the diagnostic value. RESULTS Forty-five studies with 2,843 patients and 4,263 lesions were identified. Network meta-analysis reveals that 68Ga-labeled prostate membrane antigen (68Ga-PSMA) PET/CT has the highest superiority index (7.30) with the sensitivity of 0.91 and specificity of 0.99, followed by 18F-NaF, 11C-choline, 18F-choline, 18F-fludeoxyglucose (FDG), and 18F-fluciclovine PET/CT. The use of high magnetic field strength, multisequence, diffusion-weighted imaging (DWI), and more imaging planes will increase the diagnostic value of MRI for the detection of bone metastasis in prostate cancer patients. Where available, 3.0-T high-quality MRI approaches 68Ga-PSMA PET/CT was performed in the detection of bone metastasis on patient-based level (sensitivity, 0.94 vs. 0.91; specificity, 0.94 vs. 0.96; superiority index, 4.43 vs. 4.56). CONCLUSIONS 68Ga-PSMA PET/CT is recommended for the diagnosis of bone metastasis in prostate cancer patients. Where available, 3.0-T high-quality MRI approaches 68Ga-PSMA PET/CT should be performed in the detection of bone metastasis.
Collapse
Affiliation(s)
- Fanxiao Liu
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jinlei Dong
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yelong Shen
- Department of Medical Imaging, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Canhua Yun
- Department of Nuclear Medicine, The Second Hospital of Shandong University, Jinan, China
| | - Ruixiao Wang
- Department of Urology Surgery, University Hospital of Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Ganggang Wang
- Department of Urology Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Jiyang Tan
- Department of Sports Medicine, Wuxi 9th People’s Hospital Affiliated to Soochow University, Wuxi, China
| | - Tao Wang
- Department of Sports Medicine, Wuxi 9th People’s Hospital Affiliated to Soochow University, Wuxi, China
| | - Qun Yao
- Department of Sports Medicine, Wuxi 9th People’s Hospital Affiliated to Soochow University, Wuxi, China
| | - Bomin Wang
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Lianxin Li
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jingyi Mi
- Department of Sports Medicine, Wuxi 9th People’s Hospital Affiliated to Soochow University, Wuxi, China
| | - Dongsheng Zhou
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Fei Xiong
- Department of Sports Medicine, Wuxi 9th People’s Hospital Affiliated to Soochow University, Wuxi, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
| |
Collapse
|
38
|
Sun L, Gai Y, Li Z, Zhang X, Li J, Ma Y, Li H, Barajas RJ, Zeng D. Development of Dual Receptor Enhanced Pre-Targeting Strategy-A Novel Promising Technology for Immuno-Positron Emission Tomography Imaging. ADVANCED THERAPEUTICS 2021; 4:2100110. [PMID: 35309962 PMCID: PMC8932640 DOI: 10.1002/adtp.202100110] [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: 05/17/2021] [Indexed: 11/06/2022]
Abstract
PET imaging has become an important diagnostic tool in the era of precise medicine. Various pre-targeting systems have been reported to address limitations associated with traditional immuno-PET. However, the application of these mono-receptor based pre-targeting (MRPT) strategies is limited to non-internalizable antibodies, and the tumor uptake is usually much lower than that in the corresponding immuno-PET. To circumvent these limitations, we develop the first Dual-Receptor Pre-Targeting (DRPT) system through entrapping the tumor-receptor-specific radioligand by the pre-administered antibody. Besides the similar ligation pathway happens in MRPT, incorporation of a tumor-receptor-specific peptide into the radioligand in DRPT enhances both concentration and retention of the radioligand on tumor, promoting its ligation with pre-administered mAb on cell-surface and/or internalized into tumor-cells. In this study, 64Cu based DRPT shows superior performance over corresponding MRPT and immuno-PET using internalizable antibodies. Besides, the compatibility of DRPT with short-lived and generator-produced 68Ga is demonstrated, leveraging its advantage in reducing radio-dose exposure. Furthermore, the feasibility of reducing the amount of the pre-administered antibody is confirmed, indicating the cost saving potential of DRPT. In summary, synergizing advantages of dual-receptor targeting and pre-targeting, we expect that this DRPT strategy can become a breakthrough technology in the field of antibody-based molecular imaging.
Collapse
Affiliation(s)
- Lingyi Sun
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA; Center of Radiochemistry Research, Knight Cardiovascular Institute, Oregon Health & Science University, Portland 97239, USA
| | - Yongkang Gai
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA
| | - Zhonghan Li
- Center of Radiochemistry Research, Knight Cardiovascular Institute, Oregon Health & Science University, Portland 97239, USA
| | - Xiaohui Zhang
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA
| | - Jianchun Li
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA
| | - Yongyong Ma
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA
| | - Huiqiang Li
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA
| | - Ramon J Barajas
- Department of Diagnostic Radiology, Oregon Health & Science University, Portland 97239, USA; Advanced Imaging Research Center, Oregon Health & Science University, Portland 97239, USA; Translational Oncology Research Program, Knight Cancer Institute, Oregon Health & Science University, Portland 97239, USA
| | - Dexing Zeng
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA; Center of Radiochemistry Research, Knight Cardiovascular Institute, Oregon Health & Science University, Portland 97239, USA; Department of Diagnostic Radiology, Oregon Health & Science University, Portland 97239, USA
| |
Collapse
|
39
|
Yang Z, Xie Y, Liu C, Liu X, Song S, Zhang Y, Ge R, Wang B, Yang Z. The clinical value of 18F-fluoroestradiol in assisting individualized treatment decision in dual primary malignancies. Quant Imaging Med Surg 2021; 11:3956-3965. [PMID: 34476181 DOI: 10.21037/qims-20-1364] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 04/19/2021] [Indexed: 12/29/2022]
Abstract
Background For patients with previously diagnosed dual primary tumors, it is usually difficult to determine the diagnosis and treatment of stage IV recurrence. The study was to explore the influences of 18F-fluoroestradiol positron emission tomography/computed tomography (18F-FES PET/CT) in the diagnosis of estrogen receptor (ER) positive breast cancer combined with other primary tumor with distant metastases. Methods Multidisciplinary team were organized to explore the definite clinical value of 18F-FES PET/CT in stage IV patients suffered from ER-positive breast cancer and another primary tumor synchronously or metachronously. Thirty-two female patients were retrospectively analyzed who underwent 18F-FES PET/CT scans in our center. Before and after reading 18F-FES reports, the team members from department of surgery, oncology and radiotherapy should make decisions of management strategy. Results Totally, the multidisciplinary team completed the management decision-making of the 32 patients before and after 18F-FES PET/CT scans. 87.5% (n=28) of the patients were considered to benefit from 18F-FES reports for diagnosis and treatment decisions. Out of the 28 patients, 7 patients (7/32, 21.9%) were considered to definitely change the management strategies while 12 patients (12/32, 37.5%) was instructive to develop management plans after the scan. The other 9 patients were suggested reassuring decision-making process by 18F-FES PET/CT. Conclusions 18F-FES PET/CT scans have clinical effects on diagnosis and treatment strategies of stage IV patients suffered from ER-positive breast cancer and another primary tumor.
Collapse
Affiliation(s)
- Ziyi Yang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China.,Center for Biomedical Imaging, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
| | - Yizhao Xie
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cheng Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China.,Center for Biomedical Imaging, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
| | - Xin Liu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China.,Center for Biomedical Imaging, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
| | - Yingjian Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China.,Center for Biomedical Imaging, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
| | - Rui Ge
- Department of General Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Biyun Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhongyi Yang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China.,Center for Biomedical Imaging, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
| |
Collapse
|
40
|
Boers J, Loudini N, Brunsch CL, Koza SA, de Vries EFJ, Glaudemans AWJM, Hospers GAP, Schröder CP. Value of 18F-FES PET in Solving Clinical Dilemmas in Breast Cancer Patients: A Retrospective Study. J Nucl Med 2021; 62:1214-1220. [PMID: 33990400 DOI: 10.2967/jnumed.120.256826] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 12/28/2020] [Indexed: 11/16/2022] Open
Abstract
Breast cancer (BC) is a heterogeneous disease in which estrogen receptor (ER) expression plays an important role in most tumors. A clinical dilemma may arise when a metastasis biopsy to determine the ER status cannot be performed safely or when ER heterogeneity is suspected between tumor lesions. Whole-body ER imaging, such as 16α-18F-fluoro-17β-estradiol (18F-FES) PET, may have added value in these situations. However, the role of this imaging technique in routine clinical practice remains to be further determined. Therefore, we assessed whether the physician's remaining clinical dilemma after the standard workup was solved by the 18F-FES PET scan. Methods: This retrospective study included 18F-FES PET scans of patients who had (or were suspected to have) ER-positive metastatic BC and for whom a clinical dilemma remained after the standard workup. The scans were performed at the University Medical Center of Groningen between November 2009 and January 2019. We investigated whether the physician's clinical dilemma was solved, defined either as solving the clinical dilemma through the 18F-FES PET results or as basing a treatment decision directly on the 18F-FES PET results. In addition, the category of the clinical dilemma was reported, as well as the rate of 18F-FES-positive or -negative PET scans, and any correlation to the frequency of solved dilemmas was determined. Results: One hundred 18F-FES PET scans were performed on 83 patients. The clinical dilemma categories were inability to determine the extent of metastatic disease or suspected metastatic disease with the standard workup (n = 52), unclear ER status of the tumor (n = 31), and inability to determine which primary tumor caused the metastases (n = 17). The dilemmas were solved by 18F-FES PET in 87 of 100 scans (87%). In 81 of 87 scans, a treatment decision was based directly on 18F-FES PET results (treatment change, 51 scans; continuance, 30 scans). The frequency of solved dilemmas was not related to the clinical dilemma category (P = 0.334). However, the frequency of solved dilemmas was related to whether scans were 18F-FES-positive (n = 63) or 18F-FES-negative (n = 37; P < 0.001). Conclusion: For various indications, the 18F-FES PET scan can help to solve most clinical dilemmas that may remain after the standard workup. Therefore, the 18F-FES PET scan has added value in BC patients who present the physician with a clinical dilemma.
Collapse
Affiliation(s)
- Jorianne Boers
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and
| | - Naila Loudini
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and
| | - Celina L Brunsch
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and
| | - Sylvia A Koza
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and
| | - Erik F J de Vries
- Department of Nuclear Medicine and Molecular Imaging, 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
| | - Geke A P Hospers
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and
| | - Carolina P Schröder
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and
| |
Collapse
|
41
|
Abstract
Imaging plays an integral role in the clinical care of patients with breast cancer. This review article focuses on the use of PET imaging for breast cancer, highlighting the clinical indications and limitations of 2-deoxy-2-[18F]fluoro-d-glucose (FDG) PET/CT, the potential use of PET/MRI, and 16α-[18F]fluoroestradiol (FES), a newly approved radiopharmaceutical for estrogen receptor imaging.
Collapse
Affiliation(s)
- Amy M Fowler
- Breast Imaging and Intervention Section, Department of Radiology, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI 53792-3252, USA; Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue, Madison, WI 53705, USA; University of Wisconsin Carbone Cancer Center, 600 Highland Avenue, Madison, WI 53792, USA.
| | - Steve Y Cho
- University of Wisconsin Carbone Cancer Center, 600 Highland Avenue, Madison, WI 53792, USA; Nuclear Medicine and Molecular Imaging Section, Department of Radiology, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI 53792-3252, USA
| |
Collapse
|
42
|
Hao W, Li Y, Du B, Li X. Heterogeneity of estrogen receptor based on 18F-FES PET imaging in breast cancer patients. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-021-00456-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
43
|
Heterogeneity of bone metastases as an important prognostic factor in patients affected by oestrogen receptor-positive breast cancer. The role of combined [18F]Fluoroestradiol PET/CT and [18F]Fluorodeoxyglucose PET/CT. Eur J Radiol 2021; 141:109821. [PMID: 34139575 DOI: 10.1016/j.ejrad.2021.109821] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 11/20/2022]
Abstract
PURPOSE To assess the prognostic role of different inter and intralesional expression (heterogeneity) of oestrogen receptor (ER) in bone metastases, as identified by the combined use of [18F]FES PET/CT and [18F]FDG PET/CT in patients with oestrogen receptor-positive (ER+) metastatic breast cancer (BC). METHODS We analysed patients with a new diagnosis of bone metastases who were candidates for first-line systemic endocrine therapy. Before starting therapy, patients underwent baseline [18F]FES PET/CT and [18]FDG PET/CT. Semi-quantitative evaluation of whole-body bone metabolic burden (WB-B-MB) was performed on [18F]FES and [18F]FDG PET/CT in order to evaluate disease extent, tumour metabolism and ER heterogeneity. We used time-to-event analyses (Kaplan-Meier and Cox proportional-hazards methods) to estimate progression-free (PFS) and overall survival (OS), in order to assess the independent prognostic value of [18F]FES PET/CT and [18F]FDG PET/CT, alone and in combination. RESULTS According to our criteria, we enrolled 49 patients. Over a median follow-up of 44.7 months, 35 patients suffered disease progression (71.4 %) and 15 died of disease (30.6 %). When the risk of disease progression was calculated by means of the Cox model, only [18F]FDG WB-B-MB was independently and directly associated to PFS (p = 0.02). On analysing the association between all prognostic parameters and survival, the Cox model showed that the only parameter associated with OS was the WB-B-MB FES/FDG ratio (p = 0.01). CONCLUSION The combined use of [18F]FES-PET/CT and [18F]FDG-PET/CT can identify ER heterogeneity in BC bone metastases. This heterogeneity is significantly associated with survival. Moreover, the extension of the FDG-avid component correlates with the risk of disease progression.
Collapse
|
44
|
Chaudhari AJ, Badawi RD. Application-specific nuclear medical in vivoimaging devices. Phys Med Biol 2021; 66:10TR01. [PMID: 33770765 DOI: 10.1088/1361-6560/abf275] [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: 11/05/2019] [Accepted: 03/26/2021] [Indexed: 11/11/2022]
Abstract
Nuclear medical imaging devices, such as those enabling photon emission imaging (gamma camera, single photon emission computed tomography, or positron emission imaging), that are typically used in today's clinics are optimized for assessing large portions of the human body, and are classified as whole-body imaging systems. These systems have known limitations for organ imaging, therefore application-specific devices have been designed, constructed and evaluated. These devices, given their compact nature and superior technical characteristics, such as their higher detection sensitivity and spatial resolution for organ imaging compared to whole-body imaging systems, have shown promise for niche applications. Several of these devices have further been integrated with complementary anatomical imaging devices. The objectives of this review article are to (1) provide an overview of such application-specific nuclear imaging devices that were developed over the past two decades (in the twenty-first century), with emphasis on brain, cardiac, breast, and prostate imaging; and (2) discuss the rationale, advantages and challenges associated with the translation of these devices for routine clinical imaging. Finally, a perspective on the future prospects for application-specific devices is provided, which is that sustained effort is required both to overcome design limitations which impact their utility (where these exist) and to collect the data required to define their clinical value.
Collapse
Affiliation(s)
- Abhijit J Chaudhari
- Department of Radiology, University of California Davis, Sacramento, CA 95817, United States of America
- Center for Molecular and Genomic Imaging, University of California Davis, Davis, CA 95616, United States of America
| | - Ramsey D Badawi
- Department of Radiology, University of California Davis, Sacramento, CA 95817, United States of America
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, United States of America
| |
Collapse
|
45
|
Roze JF, van Meurs HS, Monroe GR, Veldhuis WB, van Lonkhuijzen LRCW, Bennink RJ, Groeneweg JW, Witteveen PO, Jonges GN, Zweemer RP, Braat AJAT. [ 18F]FDG and [ 18F]FES positron emission tomography for disease monitoring and assessment of anti-hormonal treatment eligibility in granulosa cell tumors of the ovary. Oncotarget 2021; 12:665-673. [PMID: 33868587 PMCID: PMC8021033 DOI: 10.18632/oncotarget.27925] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/15/2021] [Indexed: 01/08/2023] Open
Abstract
Purpose: Adult granulosa cell tumors (AGCTs) of the ovary represent a rare malignancy in which timing and choice of treatment is a clinical challenge. This study investigates the value of FDG-PET/CT and FES-PET/CT in monitoring recurrent AGCTs and assessing eligibility for anti-hormonal treatment. Materials and Methods: We evaluated 22 PET/CTs from recurrent AGCT patients to determine tumor FDG (n = 16) and FES (n = 6) uptake by qualitative and quantitative analysis. We included all consecutive patients from two tertiary hospitals between 2003-2020. Expression of ERα and ERβ and mitoses per 2 mm2 were determined by immunohistochemistry and compared to FES and FDG uptake, respectively. Results: Qualitative assessment showed low-to-moderate FDG uptake in most patients (14/16), and intense uptake in 2/16. One patient with intense tumor FDG uptake had a high mitotic rate (18 per 2 mm2) Two out of six patients showed FES uptake on PET/CT at qualitative analysis. Lesion-based quantitative assessment showed a mean SUVmax of 2.4 (± 0.9) on FDG-PET/CT and mean SUVmax of 1.7 (± 0.5) on FES-PET/CT. Within patients, expression of ERα and ERβ varied and did not seem to correspond with FES uptake. In one FES positive patient, tumor locations with FES uptake remained stable or decreased in size during anti-hormonal treatment, while all FES negative locations progressed. Conclusions: This study shows that in AGCTs, FDG uptake is limited and therefore FDG-PET/CT is not advised. FES-PET/CT may be useful to non-invasively capture the estrogen receptor expression of separate tumor lesions and thus assess the potential eligibility for hormone treatment in AGCT patients.
Collapse
Affiliation(s)
- Joline F Roze
- Department of Gynecological Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Hannah S van Meurs
- Department of Gynecological Oncology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Glen R Monroe
- Department of Gynecological Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Wouter B Veldhuis
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Luc R C W van Lonkhuijzen
- Department of Gynecological Oncology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Roel J Bennink
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Jolijn W Groeneweg
- Department of Gynecological Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Petronella O Witteveen
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Geertruida N Jonges
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ronald P Zweemer
- Department of Gynecological Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Arthur J A T Braat
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
46
|
Boers J, Schröder CP, Hospers GAP, de Vries EFJ, Glaudemans AWJM. Detection of Dural Metastases Before the Onset of Clinical Symptoms by 16α-[18F]Fluoro-17β-Estradiol PET in a Patient With Estrogen Receptor-Positive Breast Cancer. Clin Nucl Med 2021; 46:e165-e167. [PMID: 33181752 DOI: 10.1097/rlu.0000000000003382] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
ABSTRACT We offer an illustrative case about estrogen receptor (ER) imaging (also known as 16α-[18F]fluoro-17β-estradiol ([18F]-FES) PET) and the detection of metastatic lesions in the dural region. We present a case of a woman with ER-positive metastatic breast cancer and high [18F]-FES uptake in the dural region on PET imaging, without associated clinical symptoms. These lesions were missed on [18F]-FDG PET because of physiological [18F]-FDG uptake in the brain. This case highlighted some difficulties in the interpretation of imaging of brain metastases and demonstrated the added value of [18F]-FES PET imaging. [18F]-FES PET could be used to prove the presence of ER-positive metastases in the brain.
Collapse
Affiliation(s)
| | | | | | - Erik F J de Vries
- Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Andor W J M Glaudemans
- Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
47
|
Su Y, Zhang Y, Hua X, Huang J, Bi X, Xia W, Wang X, Huang Z, Song C, Zhong Y, Shi Y, Wang S, Fan W, Yuan Z. High-dose tamoxifen in high-hormone-receptor-expressing advanced breast cancer patients: a phase II pilot study. Ther Adv Med Oncol 2021; 13:1758835921993436. [PMID: 33737962 PMCID: PMC7934038 DOI: 10.1177/1758835921993436] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/02/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Tumor progression following endocrine therapy is considered to indicate resistance to endocrine drugs due to a variety of mechanisms. An insufficient dose of endocrine drugs is one of the causes for treatment failure in some patients with high hormone-receptor (HR)-expressing advanced breast cancer. This study aimed to explore the efficacy of high-dose tamoxifen (TAM) treatment in patients with advanced breast cancer with highly expressed HR. MATERIALS & METHODS This was a single-arm, phase II pilot study that enrolled patients with advanced breast cancer with high HR expression (estrogen receptor ⩾60% and/or progesterone receptor ⩾60%) following routine endocrine therapy. All enrolled patients received a high-dose of TAM (100 mg/day) until disease progression. The primary endpoint was progression-free survival (PFS). The secondary endpoints included objective response rate (ORR), clinical benefit rate (CBR), overall survival (OS), and safety. Exploratory endpoints included the predictive value of 16α-18F-17β-fluoroestradiol quantitative positron emission tomography/computed tomography (18F-FES PET/CT) for treatment efficacy. RESULTS A total of 30 patients were enrolled between September 2017 and February 2019. The median PFS was 6 months [95% confidence interval (CI) 4.9-7.1] and the median OS was 15.6 months (95% CI 8.3-22.9). Five patients experienced a partial response (PR) and none experienced a complete response (CR), with an ORR of 16.7% and CBR of 33.3%. No severe adverse events were observed. Lesions with 18F-FES maximum standardized uptake value (SUVmax) ⩾4 had a significantly longer PFS [median 9.2 months, (95% CI 6.9-11.6)] compared with lesions with a 18F-FES SUVmax <4 [median 4.8 months, (95% CI 3.9-5.6); p = 0.022]. CONCLUSION A high-dose of TAM is effective and safe for patients with advanced breast cancer with high HR expression. 18F-FES SUVmax values may predict the local clinical benefits of high-dose TAM . TRIAL REGISTRATION [ClinicalTrials.gov identifier: NCT0304565].
Collapse
Affiliation(s)
- Yanhong Su
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Yarui Zhang
- Department of Nuclear Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xin Hua
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Jiajia Huang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Xiwen Bi
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Wen Xia
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Xinyue Wang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Zhangzan Huang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Chenge Song
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Yongyi Zhong
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Yanxia Shi
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Shusen Wang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | | | - Wei Fan
- Department of Nuclear Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510000, China
| | - Zhongyu Yuan
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510000, China
| |
Collapse
|
48
|
Molecular Biomarkers for Contemporary Therapies in Hormone Receptor-Positive Breast Cancer. Genes (Basel) 2021; 12:genes12020285. [PMID: 33671468 PMCID: PMC7922594 DOI: 10.3390/genes12020285] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023] Open
Abstract
Systemic treatment of hormone receptor-positive (HR+) breast cancer is undergoing a renaissance, with a number of targeted therapies including CDK4/6, mTOR, and PI3K inhibitors now approved for use in combination with endocrine therapies. The increased use of targeted therapies has changed the natural history of HR+ breast cancers, with the emergence of new escape mechanisms leading to the inevitable progression of disease in patients with advanced cancers. The identification of new predictive and pharmacodynamic biomarkers to current standard-of-care therapies and discovery of new therapies is an evolving and urgent clinical challenge in this setting. While traditional, routinely measured biomarkers such as estrogen receptors (ERs), progesterone receptors (PRs), and human epidermal growth factor receptor 2 (HER2) still represent the best prognostic and predictive biomarkers for HR+ breast cancer, a significant proportion of patients either do not respond to endocrine therapy or develop endocrine resistant disease. Genomic tests have emerged as a useful adjunct prognostication tool and guide the addition of chemotherapy to endocrine therapy. In the treatment-resistant setting, mutational profiling has been used to identify ESR1, PIK3CA, and AKT mutations as predictive molecular biomarkers to newer therapies. Additionally, pharmacodynamic biomarkers are being increasingly used and considered in the metastatic setting. In this review, we summarise the current state-of-the-art therapies; prognostic, predictive, and pharmacodynamic molecular biomarkers; and how these are impacted by emerging therapies for HR+ breast cancer.
Collapse
|
49
|
Szucs Z, Joseph J, Larkin TJ, Xie B, Bohndiek SE, Brindle KM, Neves AA. Multi-modal imaging of high-risk ductal carcinoma in situ of the breast using C2Am: a targeted cell death imaging agent. Breast Cancer Res 2021; 23:25. [PMID: 33596961 PMCID: PMC7891030 DOI: 10.1186/s13058-021-01404-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 02/01/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Ductal carcinoma in situ (DCIS) is a non-invasive form of early breast cancer, with a poorly understood natural history of invasive transformation. Necrosis is a well-recognized adverse prognostic feature of DCIS, and non-invasive detection of its presence and spatial extent could provide information not obtainable by biopsy. We describe here imaging of the distribution and extent of comedo-type necrosis in a model of human DCIS using C2Am, an imaging agent that binds to the phosphatidylserine exposed by necrotic cells. METHODS We used an established xenograft model of human DCIS that mimics the histopathological features of the disease. Planar near-infrared and optoacoustic imaging, using fluorescently labeled C2Am, were used to image non-invasively the presence and extent of lesion necrosis. RESULTS C2Am showed specific and sensitive binding to necrotic areas in DCIS tissue, detectable both in vivo and ex vivo. The imaging signal generated in vivo using near-infrared (NIR) fluorescence imaging was up to 6-fold higher in DCIS lesions than in surrounding fat pad or skin tissue. There was a correlation between the C2Am NIR fluorescence (Pearson R = 0.783, P = 0.0125) and optoacoustic signals (R > 0.875, P < 0.022) in the DCIS lesions in vivo and the corresponding levels of cell death detected histologically. CONCLUSIONS C2Am is a targeted multi-modal imaging agent that could complement current anatomical imaging methods for detecting DCIS. Imaging the presence and spatial extent of necrosis may give better prognostic information than that obtained by biopsy alone.
Collapse
Affiliation(s)
- Zoltan Szucs
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - James Joseph
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
- Department of Physics, University of Cambridge, Cambridge, UK
- Present address: University of Dundee, School of Science and Engineering, Dundee, UK
| | - Tim J Larkin
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Bangwen Xie
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Sarah E Bohndiek
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
- Department of Physics, University of Cambridge, Cambridge, UK
| | - Kevin M Brindle
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK.
- Department of Biochemistry, University of Cambridge, Cambridge, UK.
| | - André A Neves
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK.
| |
Collapse
|
50
|
Cheng X, Yang Z, Sun Y, Zheng W, Chen H, Liu Y, Wu Z. Synthesis and preliminary evaluation of a PET-FI bimodal imaging agent targeting estrogen receptor. Bioorg Med Chem Lett 2021; 34:127776. [PMID: 33418064 DOI: 10.1016/j.bmcl.2021.127776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/21/2020] [Accepted: 12/31/2020] [Indexed: 12/20/2022]
Abstract
Estrogen receptor is an attractive target for the diagnosis and treatment of breast cancer. This article reports for the first time a dual-modality imaging agent targeting estrogen receptor that can use PET imaging to diagnose breast cancer and utilize fluorescence imaging to achieve intraoperative navigation. Fluorescence experiments show that [natGa] 1 has typical aggregate induced emission characteristics. Above the critical concentration, [natGa] 1 can form biocompatible nanomicelles. [natGa] 1 can quickly light up estrogen receptor positive MCF-7 cells. Cell uptake experiments show that [68Ga] 1 is mediated by estrogen receptor. Therefore, [nat/68Ga] 1 shows the characteristics of highly sensitive diagnosis and visualization of breast cancer, and can be used as a lead compound for the development of a novel PET-FI bimodal imaging agent targeting the estrogen receptor.
Collapse
Affiliation(s)
- Xuebo Cheng
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Zequn Yang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Yuli Sun
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Wei Zheng
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Hualong Chen
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Yajing Liu
- School of Pharmaceutical Science, Capital Medical University, Beijing 100069, China.
| | - Zehui Wu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China.
| |
Collapse
|