1
|
Kitano Y, Miyake KK, Shimizu Y, Kawashima M, Nakamoto Y. Intratumoral Heterogeneity of Primary Breast Cancer on 18F-FES PET/CT and dbPET Anticipated a Heterogeneous Response to Chemotherapy. Clin Nucl Med 2025:00003072-990000000-01499. [PMID: 39848225 DOI: 10.1097/rlu.0000000000005660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
ABSTRACT A 59-year-old woman with cT3N3M1 invasive breast cancer (ER low positive, PgR positive, HER2 negative) underwent PET/CT and dedicated breast PET scans using 18F-FDG and 18F-fluoroestradiol (18F-FES). While most primary tumor regions displayed low FES uptake, regions of high FES uptake were also identified. Following chemotherapy with the paclitaxel and bevacizumab, 18F-FDG PET/CT demonstrated a favorable response, but residual disease was noted in areas with high FES uptake on the pretreatment images. This case illustrated that 18F-FES PET can depict intratumoral heterogeneity within the primary tumor, which was strongly associated with a heterogeneous response to subsequent chemotherapy.
Collapse
Affiliation(s)
- Yurika Kitano
- From the Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine Kyoto University, Kyoto, Japan
| | - Kanae K Miyake
- Department of Advanced Imaging in Medical Magnetic Resonance, Graduate School of Medicine Kyoto University, Kyoto, Japan
| | - Yoichi Shimizu
- From the Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine Kyoto University, Kyoto, Japan
| | - Masahiro Kawashima
- Department of Breast Surgery, Graduate School of Medicine Kyoto University, Kyoto, Japan
| | - Yuji Nakamoto
- From the Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine Kyoto University, Kyoto, Japan
| |
Collapse
|
2
|
Young AJ, Pantel AR, Kiani M, Doot RK, Bagheri S, Pryma DA, Farwell MD, Li S, Lee H, Schubert EK, Secreto A, Zuckerman SP, Nayak A, Choi H, Carlin S, DeMichele A, Mankoff DA, Zhou R, Mach RH, McDonald ES. Kinetic Analysis and Metabolism of Poly(Adenosine Diphosphate-Ribose) Polymerase-1-Targeted 18F-Fluorthanatrace PET in Breast Cancer. J Nucl Med 2024; 65:1862-1868. [PMID: 39477499 PMCID: PMC11619586 DOI: 10.2967/jnumed.124.268254] [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: 06/17/2024] [Accepted: 09/11/2024] [Indexed: 12/08/2024] Open
Abstract
The poly(adenosine diphosphate-ribose) polymerase inhibitors (PARPi) have demonstrated efficacy in ovarian, breast, and prostate cancers, but current biomarkers do not consistently predict clinical benefit. 18F-fluorthanatrace (18F-FTT) is an analog to rucaparib, a clinically approved PARPi, and is a candidate biomarker for PARPi response. This study intends to characterize 18F-FTT pharmacokinetics in breast cancer and optimize image timing for clinical trials. A secondary aim is to determine whether 18F-FTT uptake in breast cancer correlates with matched frozen surgical specimens as a reference standard for PARP-1 protein. Methods: Thirty prospectively enrolled women with a new diagnosis of breast cancer were injected with 18F-FTT and imaged dynamically 0-60 min after injection over the chest, with an optional static scan over multiple bed positions starting around 70 min. Kinetic analysis of lesion uptake was performed using blood-pool activity with population radiometabolite corrections. Normal breast and normal muscle reference tissue models were compared with PARP-1 protein expression in 10 patients with available tissue. Plasma radiometabolite concentrations and uptake in tumor and normal muscle were investigated in mouse xenografts. Results: Pharmacokinetics of 18F-FTT were well fit by Logan plot reference region models of reversible binding. However, fits of 2-tissue compartment models assuming negligible metabolite uptake were unstable. Rapid metabolism of 18F-FTT was demonstrated in mice, and similar uptake of radiometabolites was found in tumor xenografts and normal muscle. Tumor 18F-FTT distribution volume ratios relative to normal muscle reference tissue correlated with tissue PARP-1 expression (P < 0.02, n = 10). The tumor-to-normal muscle ratio from a 5-min frame between 50 and 60 min after injection, a potential static scan protocol, closely corresponded to the distribution volume ratio relative to normal muscle and correlated to PARP-1 expression (P < 0.02, n = 10). Conclusion: This study of PARPi analog 18F-FTT showed that uptake kinetics in vivo corresponded to expression of PARP-1 and that 18F-FTT quantitation is influenced by radiometabolites that are increasingly present late after injection. Radiometabolites can be controlled by using optimal image acquisition timing or normal muscle reference tissue modeling in dynamic imaging or a tumor-to-normal muscle ratio. Optimal image timing for tumor-to-normal muscle quantification in humans appears to be between 50 and 60 min after injection. Therefore, a clinically practical static imaging protocol commencing 45-55 min after injection may sufficiently balance 18F-FTT uptake with background clearance and radiometabolite interference for quantitative interpretation of PARP-1 expression in vivo.
Collapse
Affiliation(s)
- Anthony J Young
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Austin R Pantel
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mahsa Kiani
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert K Doot
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sina Bagheri
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniel A Pryma
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael D Farwell
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shihong Li
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hsiaoju Lee
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Erin K Schubert
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anthony Secreto
- Department of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Samantha P Zuckerman
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anupma Nayak
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hoon Choi
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Angela DeMichele
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David A Mankoff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rong Zhou
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert H Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth S McDonald
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania;
| |
Collapse
|
3
|
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
|
4
|
O'Brien SR, Edmonds CE, Ward RE, Taunk NK, Pantel AR, Mankoff DA. Update on 18F-Fluoroestradiol. Semin Nucl Med 2024; 54:812-826. [PMID: 39368910 DOI: 10.1053/j.semnuclmed.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 10/07/2024]
Abstract
18F-16α-Fluoroestradiol (18F-FES) is a radiolabeled estrogen analogue positron emission tomography (PET) imaging agent that binds to the estrogen receptor (ER) in the nucleus of ER-expressing cells. Proof-of-concept studies of 18F-FES demonstrated expected correlation between tumoral 18F-FES-positivity on PET-imaging and ER+ status assessed on biopsy samples by radioligand binding and immunohistochemistry. After decades of study, 18F-FES PET/CT gained clinical approval in 2016 in France and 2020 in the United States for use in patients with ER+ metastatic or recurrent breast cancer. ER+ as assessed by 18F-FES PET/CT has been shown to serve as a biomarker, identifying metastatic breast cancer patients who may respond to endocrine therapy and those who are unlikely to respond. In 2023, the Society of Nuclear Medicine and Molecular Imaging (SNMMI) published Appropriate Use Criteria for 18F-FES PET/CT, identifying four indications in which use of 18F-FES PET/CT was "appropriate": (1) To assess functional ER status in metastatic lesions unfavorable to biopsy or when biopsy is nondiagnostic, (2) To detect ER status when other imaging tests are equivocal or suspicious, and at (3) initial diagnosis of metastatic disease or (4) progression of metastatic disease, for considering endocrine therapy. This article reviews the foundations of 18F-FES imaging, including normal distribution, false positives, and false negatives, and describes the most up-to-date clinical uses as well as emerging research in breast cancer and other patient populations.
Collapse
Affiliation(s)
- Sophia R O'Brien
- Division of Nuclear Medicine Imaging and Therapy, Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA; Department of Radiology, Division of Breast Imaging, Hospital of the University of Pennsylvania, Philadelphia, PA.
| | - Christine E Edmonds
- Department of Radiology, Division of Breast Imaging, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Rebecca E Ward
- Division of Nuclear Medicine Imaging and Therapy, Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Neil K Taunk
- Department of Radiation Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Austin R Pantel
- Division of Nuclear Medicine Imaging and Therapy, Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - David A Mankoff
- Division of Nuclear Medicine Imaging and Therapy, Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
5
|
Binmujlli MA. Exploring Radioiodinated Anastrozole and Epirubicin as AKT1-Targeted Radiopharmaceuticals in Breast Cancer: In Silico Analysis and Potential Therapeutic Effect with Functional Nuclear Imagining Implications. Molecules 2024; 29:4203. [PMID: 39275052 PMCID: PMC11397058 DOI: 10.3390/molecules29174203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 08/29/2024] [Accepted: 09/03/2024] [Indexed: 09/16/2024] Open
Abstract
This study evaluates radio-iodinated anastrozole ([125I]anastrozole) and epirubicin ([125I]epirubicin) for AKT1-targeted breast cancer therapy, utilizing radiopharmaceutical therapy (RPT) for personalized treatment. Through molecular docking and dynamics simulations (200 ns), it investigates these compounds' binding affinities and mechanisms to the AKT1 enzyme, compared to the co-crystallized ligand, a known AKT1 inhibitor. Molecular docking results show that [125I]epirubicin has the highest ΔGbind (-11.84 kcal/mol), indicating a superior binding affinity compared to [125I] anastrozole (-10.68 kcal/mol) and the co-crystallized ligand (-9.53 kcal/mol). Molecular dynamics (MD) simulations confirmed a stable interaction with the AKT1 enzyme, with [125I]anastrozole and [125I]epirubicin reaching stability after approximately 68 ns with an average RMSD of around 2.2 Å, while the co-crystallized ligand stabilized at approximately 2.69 Å after 87 ns. RMSF analysis showed no significant shifts in residues or segments, with consistent patterns and differences of less than 2 Å, maintaining enzyme stability. The [125I]epirubicin complex maintained an average of four H-bonds, indicating strong and stable interactions, while [125I]anastrozole consistently formed three H-bonds. The average Rg values for both complexes were ~16.8 ± 0.1 Å, indicating no significant changes in the enzyme's compactness, thus preserving structural integrity. These analyses reveal stable binding and minimal structural perturbations, suggesting the high potential for AKT1 inhibition. MM-PBSA calculations confirm the potential of these radio-iodinated compounds as AKT1 inhibitors, with [125I]epirubicin exhibiting the most favorable binding energy (-23.57 ± 0.14 kcal/mol) compared to [125I]anastrozole (-20.03 ± 0.15 kcal/mol) and the co-crystallized ligand (-16.38 ± 0.14 kcal/mol), highlighting the significant role of electrostatic interactions in stabilizing the complex. The computational analysis shows [125I]anastrozole and [125I]epirubicin may play promising roles as AKT1 inhibitors, especially [125I]epirubicin for its high binding affinity and dynamic receptor interactions. These findings, supported by molecular docking scores and MM-PBSA binding energies, advocate for their potential superior inhibitory capability against the AKT1 enzyme. Nevertheless, it is crucial to validate these computational predictions through in vitro and in vivo studies to thoroughly evaluate the therapeutic potential and viability of these compounds for AKT1-targeted breast cancer treatment.
Collapse
Affiliation(s)
- Mazen Abdulrahman Binmujlli
- Department of Internal Medicine, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), P.O. Box 90950, Riyadh 11623, Saudi Arabia
| |
Collapse
|
6
|
Sluka P, Ackermann U, Rigopoulos A, Wardan H, Pezaro C, Burvenich IJ, Scott AM, Davis ID. Characterization of an Estrogen Receptor α-Selective 18 F-Estradiol PET Tracer. World J Nucl Med 2024; 23:153-160. [PMID: 39170834 PMCID: PMC11335392 DOI: 10.1055/s-0044-1786518] [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] [Indexed: 08/23/2024] Open
Abstract
Objective Conventional imaging of cancer with modalities such as computed tomography or magnetic resonance imaging provides little information about the underlying biology of the cancer and consequently little guidance for systemic treatment choices. Accurate identification of aggressive cancers or those that are likely to respond to specific treatment regimens would allow more precisely tailored treatments to be used. The expression of the estrogen receptor α subunit is associated with a more aggressive phenotype, with a greater propensity to metastasize. We aimed to characterize the binding properties of an 18 F-estradiol positron emission tomography (PET) tracer in its ability to bind to the α and β forms of estrogen receptors in vitro and confirmed its binding to estrogen receptor α in vivo. Methods The 18 F-estradiol PET tracer was synthesized and its quality confirmed by high-performance liquid chromatography. Binding of the tracer was assessed in vitro by saturation and competitive binding studies to HEK293T cells transfected with estrogen receptor α ( ESR1 ) and/or estrogen receptor β ( ESR2 ). Binding of the tracer to estrogen receptor α in vivo was assessed by imaging of uptake of the tracer into MCF7 xenografts in BALB/c nu/nu mice. Results The 18 F-estradiol PET tracer bound with high affinity (94 nM) to estrogen receptor α, with negligible binding to estrogen receptor β. Uptake of the tracer was observed in MCF7 xenografts, which almost exclusively express estrogen receptor α. Conclusion 18 F-estradiol PET tracer binds in vitro with high specificity to the estrogen receptor α isoform, with minimal binding to estrogen receptor β. This may help distinguish human cancers with biological dependence on estrogen receptor subtypes.
Collapse
Affiliation(s)
- Pavel Sluka
- Eastern Health Clinical School, Monash University, Box Hill, VIC, Australia
| | - Uwe Ackermann
- Department of Molecular Imaging and Therapy, Austin Hospital, Heidelberg, VIC, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
- The University of Melbourne, Parkville, VIC, Australia
| | - Angela Rigopoulos
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Hady Wardan
- Eastern Health Clinical School, Monash University, Box Hill, VIC, Australia
| | - Carmel Pezaro
- Eastern Health Clinical School, Monash University, Box Hill, VIC, Australia
- Department of Oncology, Eastern Health, Box Hill, VIC, Australia
| | - Ingrid J.G. Burvenich
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Andrew M. Scott
- Department of Molecular Imaging and Therapy, Austin Hospital, Heidelberg, VIC, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
- The University of Melbourne, Parkville, VIC, Australia
| | - Ian D. Davis
- Eastern Health Clinical School, Monash University, Box Hill, VIC, Australia
- Department of Oncology, Eastern Health, Box Hill, VIC, Australia
| |
Collapse
|
7
|
Lin LL, Wong F, Lin R, Yap T, Litton JK. Pharmacodynamic Activity of [ 18F]-Fluorthanatrace Poly(ADP-ribose) Polymerase Positron Emission Tomography in Patients With BRCA1/2-Mutated Breast Cancer Receiving Talazoparib. JCO Precis Oncol 2024; 8:e2400303. [PMID: 39208372 DOI: 10.1200/po.24.00303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/08/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
PURPOSE We tested the ability of [18F] fluorthanatrace (FTT), a radiolabeled analog of poly(ADP-ribose) polymerase (PARP)-1 inhibitors, to demonstrate target engagement on positron emission tomography (PET) scans from patients with newly diagnosed primary breast cancer receiving the PARP inhibitor (PARPi) talazoparib. METHODS Seven patients with germline BRCA1/2 pathogenic variants underwent [18F]FTT PET-computed tomography scanning at baseline, and five underwent repeat scanning 14 days after talazoparib initiation. Maximum uptake on PET was quantified in the primary tumor, involved nodes, contralateral pectoralis muscle, and lumbar vertebra body level 3, and compared between the two time points. RESULTS Blocking of [18F]FTT was observed on the second scan. Potentially strong but nonsignificant correlations were found between changes in tumor volume (on ultrasound at 1 month v baseline) and percentage changes in tumor-to-muscle uptake ratio at 14 days from baseline (Spearman rank correlation coefficient r = 1; P = .083); and between the highest-grade hematologic toxicity and baseline bone marrow-to-muscle (B/M) uptake ratio (r = 0.72; P = .068) and percentage change in B/M ratio at 14 days from baseline (r = 0.87; P = .058). CONCLUSION We conclude that [18F]FTT can image target engagement by PARPi, but larger studies are needed to determine whether [18F]FTT uptake can predict response to PARPi and whether uptake of [18F]FTT in bone marrow may be an early predictor of hematologic toxicity.
Collapse
Affiliation(s)
- Lilie L Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Franklin Wong
- Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ruitao Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Timothy Yap
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX
- Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jennifer K Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
8
|
Liu C, Ma G, Xu X, Song S, Yang Z. Can 18F-FES PET Improve the Evaluation of 18F-FDG PET in Patients With Metastatic Invasive Lobular Carcinoma? Clin Nucl Med 2024; 49:301-307. [PMID: 38427956 DOI: 10.1097/rlu.0000000000005085] [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: 03/03/2024]
Abstract
PURPOSE Invasive lobular carcinoma (ILC) exhibits a low affinity for 18F-FDG. The estrogen receptor (ER) is commonly expressed in ILCs, suggesting a potential benefit of targeting with the ER probe 18F-FES in this patient population. The objective of this study was to evaluate the diagnostic performance of 18F-FES imaging in patients with metastatic ILC and compare it with that of 18F-FDG. METHODS We conducted a retrospective analysis of 20 ILC patients who underwent concurrent 18F-FES and 18F-FDG PET/CT examinations in our center. 18F-FES and 18F-FDG imaging were analyzed to determine the total count of tracer-avid lesions in nonbone sites and their corresponding organ systems, assess the extent of anatomical regions involved in bone metastases, and measure the SUVmax values for both tracers. RESULTS Among 20 ILC patients, 65 nonbone lesions were found to be distributed in 13 patients, and 16 patients were diagnosed with bone metastasis, which was distributed in 54 skeletal anatomical regions. The detection rate of 18F-FDG in nonbone lesions was higher than that of 18F-FES (57 vs 37, P < 0.001). 18F-FES demonstrated a superior ability to detect nonbone lesions in 4 patients, whereas 18F-FDG was superior in 5 patients (P > 0.05). Among 9/16 patients with bone metastasis, 18F-FES demonstrated a significant advantage in the detection of bone lesions compared with 18F-FDG (P = 0.05). Furthermore, patients with only 18F-FES-positive lesions (12/12) were administered endocrine regimens, whereas patients lacking 18F-FES uptake (2/3) predominantly received chemotherapy. CONCLUSIONS 18F-FES is more effective than 18F-FDG in detecting bone metastasis in ILC, but it does not demonstrate a significant advantage in nonbone lesions. Additionally, the results of examination with 18F-FES have the potential to guide patient treatment plans.
Collapse
|
9
|
Pedersen MA, Munk OL, Dias AH, Steffensen JH, Møller AL, Johnsson AL, Hansen KV, Bender D, Jakobsen S, Busk M, Gormsen LC, Tramm T, Borgquist S, Vendelbo MH. Dynamic whole-body [ 18F]FES PET/CT increases lesion visibility in patients with metastatic breast cancer. EJNMMI Res 2024; 14:24. [PMID: 38436824 PMCID: PMC10912074 DOI: 10.1186/s13550-024-01080-y] [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: 01/10/2024] [Accepted: 02/15/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Correct classification of estrogen receptor (ER) status is essential for prognosis and treatment planning in patients with breast cancer (BC). Therefore, it is recommended to sample tumor tissue from an accessible metastasis. However, ER expression can show intra- and intertumoral heterogeneity. 16α-[18F]fluoroestradiol ([18F]FES) Positron Emission Tomography/Computed Tomography (PET/CT) allows noninvasive whole-body (WB) identification of ER distribution and is usually performed as a single static image 60 min after radiotracer injection. Using dynamic whole-body (D-WB) PET imaging, we examine [18F]FES kinetics and explore whether Patlak parametric images ( K i ) are quantitative and improve lesion visibility. RESULTS This prospective study included eight patients with metastatic ER-positive BC scanned using a D-WB PET acquisition protocol. The kinetics of [18F]FES were best characterized by the irreversible two-tissue compartment model in tumor lesions and in the majority of organ tissues. K i values from Patlak parametric images correlated with K i values from the full kinetic analysis, r2 = 0.77, and with the semiquantitative mean standardized uptake value (SUVmean), r2 = 0.91. Furthermore, parametric K i images had the highest target-to-background ratio (TBR) in 162/164 metastatic lesions and the highest contrast-to-noise ratio (CNR) in 99/164 lesions compared to conventional SUV images. TBR was 2.45 (95% confidence interval (CI): 2.25-2.68) and CNR 1.17 (95% CI: 1.08-1.26) times higher in K i images compared to SUV images. These quantitative differences were seen as reduced background activity in the K i images. CONCLUSION [18F]FES uptake is best described by an irreversible two-tissue compartment model. D-WB [18F]FES PET/CT scans can be used for direct reconstruction of parametric K i images, with superior lesion visibility and K i values comparable to K i values found from full kinetic analyses. This may aid correct ER classification and treatment decisions. Trial registration ClinicalTrials.gov: NCT04150731, https://clinicaltrials.gov/study/NCT04150731.
Collapse
Affiliation(s)
- Mette A Pedersen
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Palle-Juul-Jensens Boulevard 165, 8200, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Ole L Munk
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Palle-Juul-Jensens Boulevard 165, 8200, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - André H Dias
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Palle-Juul-Jensens Boulevard 165, 8200, Aarhus, Denmark
| | | | - Anders L Møller
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Kim Vang Hansen
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Palle-Juul-Jensens Boulevard 165, 8200, Aarhus, Denmark
| | - Dirk Bender
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Palle-Juul-Jensens Boulevard 165, 8200, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Steen Jakobsen
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Palle-Juul-Jensens Boulevard 165, 8200, Aarhus, Denmark
| | - Morten Busk
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| | - Lars C Gormsen
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Palle-Juul-Jensens Boulevard 165, 8200, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Trine Tramm
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Signe Borgquist
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Mikkel H Vendelbo
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Palle-Juul-Jensens Boulevard 165, 8200, Aarhus, Denmark.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
10
|
Kalaba P, Sanchez de la Rosa C, Möller A, Alewood PF, Muttenthaler M. Targeting the Oxytocin Receptor for Breast Cancer Management: A Niche for Peptide Tracers. J Med Chem 2024; 67:1625-1640. [PMID: 38235665 PMCID: PMC10859963 DOI: 10.1021/acs.jmedchem.3c01089] [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: 06/15/2023] [Revised: 12/07/2023] [Accepted: 12/20/2023] [Indexed: 01/19/2024]
Abstract
Breast cancer is a leading cause of death in women, and its management highly depends on early disease diagnosis and monitoring. This remains challenging due to breast cancer's heterogeneity and a scarcity of specific biomarkers that could predict responses to therapy and enable personalized treatment. This Perspective describes the diagnostic landscape for breast cancer management, molecular strategies targeting receptors overexpressed in tumors, the theranostic potential of the oxytocin receptor (OTR) as an emerging breast cancer target, and the development of OTR-specific optical and nuclear tracers to study, visualize, and treat tumors. A special focus is on the chemistry and pharmacology underpinning OTR tracer development, preclinical in vitro and in vivo studies, challenges, and future directions. The use of peptide-based tracers targeting upregulated receptors in cancer is a highly promising strategy complementing current diagnostics and therapies and providing new opportunities to improve cancer management and patient survival.
Collapse
Affiliation(s)
- Predrag Kalaba
- Institute
of Biological Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | | | - Andreas Möller
- QIMR
Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- The
Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Paul F. Alewood
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, Queensland 4072, Australia
| | - Markus Muttenthaler
- Institute
of Biological Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
11
|
Ali MU, Chaudhary BN, Panja S, Gendelman HE. Theranostic Diagnostics. Results Probl Cell Differ 2024; 73:551-578. [PMID: 39242393 DOI: 10.1007/978-3-031-62036-2_22] [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: 09/09/2024]
Abstract
Diagnosing and then treating disease defines theranostics. The approach holds promise by facilitating targeted disease outcomes. The simultaneous analysis of finding the presence of disease pathophysiology while providing a parallel in treatment is a novel and effective strategy for seeking improved medical care. We discuss how theranostics improves disease outcomes is discussed. The chapter reviews the delivery of targeted therapies. Bioimaging techniques are highlighted as early detection and tracking systems for microbial infections, degenerative diseases, and cancers.
Collapse
Affiliation(s)
- Mohammad Uzair Ali
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bharat N Chaudhary
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sudipta Panja
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
12
|
Kiatkittikul P, Mayurasakorn S, Promteangtrong C, Kunawudhi A, Siripongsatian D, Hirata N, Jantarato A, Boonkawin N, Yaset S, Kongsakorn P, Phewnual W, Chotipanich C. Head-to-head comparison of 18F-FDG and 18F-FES PET/CT for initial staging of ER-positive breast cancer patients. Eur J Hybrid Imaging 2023; 7:23. [PMID: 38105347 PMCID: PMC10725859 DOI: 10.1186/s41824-023-00176-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 08/28/2023] [Indexed: 12/19/2023] Open
Abstract
PURPOSE To compare the diagnostic performance of 18F-fluorodeoxyglucose (18F-FDG) and 18F-fluoroestradiol (18F-FES) positron emission tomography/computed tomography (PET/CT) for initial staging of estrogen receptor (ER) positive breast cancer. METHODS Twenty-eight patients with ER-positive breast cancer underwent 18F-FDG and 18F-FES PET/CT for initial staging. Diagnostic performance and concordance rates were analyzed for both radiotracers. Semiquantitative parameters of maximum standardized uptake value (SUVmax) and tumor-to-normal ratio (T/N ratio) were compared using Wilcoxon signed-rank test. Factors potentially affecting the degree of radiotracer uptake were analyzed by multi-level linear regression analysis. RESULTS The overall diagnostic performance of 18F-FES was comparable to 18F-FDG, except for higher specificity and NPV, with sensitivity, specificity, PPV, NPV, and accuracy of 87.56%, 100%, 100%, 35.14%, and 88.35%, respectively, for 18F-FES and 83.94%, 30.77%, 94.74%, 11.43%, and 95.37%, respectively, for 18F-FDG. Diagnostic performance of strong ER expression was better in 18F-FES but worse for 18F-FDG. There was a correlation of mucinous cell type and Allred score 7-8 with 18F-FES uptake, with correlation coefficients of 26.65 (19.28, 34.02), 5.90 (- 0.005, 11.81), and p-value of < 0.001, 0.05, respectively. Meanwhile, luminal B and Ki-67 were related to 18F-FDG uptake, with correlation coefficients of 2.76 (1.10, 0.20), 0.11 (0.01, 0.2), and p-value of 0.018, 0.025, respectively. CONCLUSION Diagnostic performance of 18F-FES is comparable to 18F-FDG, but better for strongly ER-positive breast cancer. Combination of 18F-FES and 18F-FDG would potentially overcome the limitations of each tracer with more accurate staging.
Collapse
Affiliation(s)
- Peerapon Kiatkittikul
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, 906 Kamphaeng Phet 6 Rd., Lak Si, Bangkok, 10210, Thailand.
| | - Supanida Mayurasakorn
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, 906 Kamphaeng Phet 6 Rd., Lak Si, Bangkok, 10210, Thailand
| | - Chetsadaporn Promteangtrong
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, 906 Kamphaeng Phet 6 Rd., Lak Si, Bangkok, 10210, Thailand
| | - Anchisa Kunawudhi
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, 906 Kamphaeng Phet 6 Rd., Lak Si, Bangkok, 10210, Thailand
| | - Dheeratama Siripongsatian
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, 906 Kamphaeng Phet 6 Rd., Lak Si, Bangkok, 10210, Thailand
| | - Natdanai Hirata
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, 906 Kamphaeng Phet 6 Rd., Lak Si, Bangkok, 10210, Thailand
| | - Attapon Jantarato
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, 906 Kamphaeng Phet 6 Rd., Lak Si, Bangkok, 10210, Thailand
| | - Natphimol Boonkawin
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, 906 Kamphaeng Phet 6 Rd., Lak Si, Bangkok, 10210, Thailand
| | - Sukanya Yaset
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, 906 Kamphaeng Phet 6 Rd., Lak Si, Bangkok, 10210, Thailand
| | - Pattanapong Kongsakorn
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, 906 Kamphaeng Phet 6 Rd., Lak Si, Bangkok, 10210, Thailand
| | - Warunya Phewnual
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, 906 Kamphaeng Phet 6 Rd., Lak Si, Bangkok, 10210, Thailand
| | - Chanisa Chotipanich
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, 906 Kamphaeng Phet 6 Rd., Lak Si, Bangkok, 10210, Thailand
| |
Collapse
|
13
|
Matushita CS, Coelho FDARFB, Stasiak CES, Rodrigues DF, Pianta DB, Kurkowski FD, da Silva MM, de Souza SAL, Lopes RW, de Castro PHR. 18F-fluoroestradiol positron emission tomography in patients with breast cancer: a systematic review and meta-analysis. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2023; 69:e2023S116. [PMID: 37556635 PMCID: PMC10411708 DOI: 10.1590/1806-9282.2023s116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 03/21/2023] [Indexed: 08/11/2023]
Affiliation(s)
- Cristina S. Matushita
- Sociedade Brasileira de Medicina Nuclear – São Paulo (SP), Brazil
- Pontifícia Universidade Católica do Rio Grande do Sul, Instituto Do Cérebro do Rio Grande do Sul – Porto Alegre (RS), Brazil
- São Marcos Diagnósticos por Imagem – Joinville (SC), Brazil
| | | | - Camila Edith Stachera Stasiak
- Universidade Federal do Rio de Janeiro, Faculdade de Medicina, Departamento de Radiologia – Rio de Janeiro (RJ), Brazil
- Universidade Federal Fluminense, Serviço de Medicina Nuclear – Niterói (RJ), Brazil
| | - Denise Ferreira Rodrigues
- Nuclear Medcenter – Belo Horizonte (MG), Brazil
- Axial Inteligência Diagnóstica – Belo Horizonte (MG), Brazil
| | - Diego Bromfman Pianta
- Sociedade Brasileira de Medicina Nuclear – São Paulo (SP), Brazil
- Pontifícia Universidade Católica do Rio Grande do Sul, Instituto Do Cérebro do Rio Grande do Sul – Porto Alegre (RS), Brazil
- São Marcos Diagnósticos por Imagem – Joinville (SC), Brazil
| | - Flávia Dornelas Kurkowski
- Pontifícia Universidade Católica do Rio Grande do Sul, Instituto Do Cérebro do Rio Grande do Sul – Porto Alegre (RS), Brazil
| | - Marcelo Moreira da Silva
- São Marcos Diagnósticos por Imagem – Joinville (SC), Brazil
- Instituto de Medicina Nuclear e Endo de Brasília – Brasília (DF), Brazil
| | - Sergio Augusto Lopes de Souza
- Universidade Federal do Rio de Janeiro, Faculdade de Medicina, Departamento de Radiologia – Rio de Janeiro (RJ), Brazil
| | - Rafael Willain Lopes
- Sociedade Brasileira de Medicina Nuclear – São Paulo (SP), Brazil
- Hospital do Coração, Setor de Medicina Nuclear – São Paulo (SP), Brazil
| | - Paulo Henrique Rosado de Castro
- Sociedade Brasileira de Medicina Nuclear – São Paulo (SP), Brazil
- Pontifícia Universidade Católica do Rio Grande do Sul, Instituto Do Cérebro do Rio Grande do Sul – Porto Alegre (RS), Brazil
- São Marcos Diagnósticos por Imagem – Joinville (SC), Brazil
| |
Collapse
|
14
|
Covington MF, Hoffman JM, Morton KA, Buckway B, Boucher KM, Rosenthal RE, Porretta JM, Brownson KE, Matsen CB, Vaklavas C, Ward JH, Wei M, Buys SS, Chittoria N, Yakish ED, Archibald ZG, Burrell LD, Butterfield RI, Yap JT. Prospective Pilot Study of 18F-Fluoroestradiol PET/CT in Patients With Invasive Lobular Carcinomas. AJR Am J Roentgenol 2023; 221:228-239. [PMID: 36919879 DOI: 10.2214/ajr.22.28809] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
BACKGROUND. PET/CT with 18F-fluoroestradiol (FES) (FDA-approved in 2020) depicts tissues expressing estrogen receptor (ER). Invasive lobular carcinoma (ILC) is commonly ER positive. OBJECTIVE. The primary aim of this study was to assess the frequency with which sites of histologically proven ILC have abnormal uptake on FES PET/CT. METHODS. This prospective single-center pilot study, conducted from December 2020 to August 2021, enrolled patients with histologically confirmed ILC to undergo FES PET/CT; patients optionally underwent FDG PET/CT. Two nuclear radiologists assessed FES PET/CT and FDG PET/CT studies for abnormal uptake corresponding to known ILC sites at enrollment and for additional sites of abnormal uptake, resolving differences by consensus. The primary endpoint was percentage of known ILC sites showing abnormal FES uptake. The alternative to the null hypothesis was that more than 60% of sites would have abnormal FES uptake, exceeding the percentage of ILC with abnormal FDG uptake described in prior literature. A sample size of 24 biopsied lesions was preselected to provide 81% power for the alternative hypothesis (one-sided α = .10). Findings on FES PET/CT and FDG PET/CT were summarized for additional secondary endpoints. RESULTS. The final analysis included 17 patients (mean age, 59.1 ± 13.2 years) with 25 sites of histologically confirmed ILC at enrollment (22 breast lesions, two axillary lymph nodes, one distant metastasis). FES PET/CT showed abnormal uptake in 22 of 25 (88%) lesions, sufficient to reject the null hypothesis (p = .002). Thirteen patients underwent FDG PET/CT. Four of 23 (17%) sites of histologically confirmed ILC, including additional sites detected and confirmed after enrollment, were identified with FES PET/CT only, and 1 of 23 (4%) was identified only with FDG PET/CT (p = .18). FES PET/CT depicted additional lesions not detected with standard-of-care evaluation in 4 of 17 (24%) patients (two contralateral breast cancers and two metastatic axillary lymph nodes, all with subsequent histologic confirmation). Use of FES PET/CT resulted in changes in clinical stage with respect to standard-of-care evaluation in 3 of 17 (18%) patients. CONCLUSION. The primary endpoint of the trial was met. The frequency of abnormal FES uptake among sites of histologically known ILC was found to be to be significantly greater than 60%. CLINICAL IMPACT. This pilot study shows a potential role of FES PET/CT in evaluation of patients with ILC. TRIAL REGISTRATION. ClinicalTrials.gov NCT04252859.
Collapse
Affiliation(s)
- Matthew F Covington
- Center for Quantitative Cancer Imaging, Huntsman Cancer Institute, 2000 Circle of Hope Dr, Salt Lake City, UT 84112
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT
| | - John M Hoffman
- Center for Quantitative Cancer Imaging, Huntsman Cancer Institute, 2000 Circle of Hope Dr, Salt Lake City, UT 84112
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT
| | - Kathryn A Morton
- Center for Quantitative Cancer Imaging, Huntsman Cancer Institute, 2000 Circle of Hope Dr, Salt Lake City, UT 84112
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT
- Present affiliation: Summit Physician Specialists, Murray, UT
| | - Brandon Buckway
- Center for Quantitative Cancer Imaging, Huntsman Cancer Institute, 2000 Circle of Hope Dr, Salt Lake City, UT 84112
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT
| | | | | | - Jane M Porretta
- Department of Surgery, University of Utah, Salt Lake City, UT
| | | | - Cindy B Matsen
- Department of Surgery, University of Utah, Salt Lake City, UT
| | - Christos Vaklavas
- Department of Internal Medicine, Oncology Division, University of Utah, Salt Lake City, UT
| | - John H Ward
- Department of Internal Medicine, Oncology Division, University of Utah, Salt Lake City, UT
| | - Mei Wei
- Department of Internal Medicine, Oncology Division, University of Utah, Salt Lake City, UT
| | - Saundra S Buys
- Department of Internal Medicine, Oncology Division, University of Utah, Salt Lake City, UT
| | - Namita Chittoria
- Department of Internal Medicine, Oncology Division, University of Utah, Salt Lake City, UT
| | - Ellen D Yakish
- Center for Quantitative Cancer Imaging, Huntsman Cancer Institute, 2000 Circle of Hope Dr, Salt Lake City, UT 84112
| | - Zane G Archibald
- Center for Quantitative Cancer Imaging, Huntsman Cancer Institute, 2000 Circle of Hope Dr, Salt Lake City, UT 84112
| | - Lance D Burrell
- Center for Quantitative Cancer Imaging, Huntsman Cancer Institute, 2000 Circle of Hope Dr, Salt Lake City, UT 84112
- Present affiliation: Society of Nuclear Medicine and Molecular Imaging, Reston, VA
| | - Regan I Butterfield
- Center for Quantitative Cancer Imaging, Huntsman Cancer Institute, 2000 Circle of Hope Dr, Salt Lake City, UT 84112
| | - Jeffrey T Yap
- Center for Quantitative Cancer Imaging, Huntsman Cancer Institute, 2000 Circle of Hope Dr, Salt Lake City, UT 84112
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
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
|
18
|
Zhang-Yin J. State of the Art in 2022 PET/CT in Breast Cancer: A Review. J Clin Med 2023; 12:968. [PMID: 36769616 PMCID: PMC9917740 DOI: 10.3390/jcm12030968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 02/01/2023] Open
Abstract
Molecular imaging with positron emission tomography is a powerful and well-established tool in breast cancer management. In this review, we aim to address the current place of the main PET radiopharmaceuticals in breast cancer care and offer perspectives on potential future radiopharmaceutical and technological advancements. A special focus is given to the following: the role of 18F-fluorodeoxyglucose positron emission tomography in the clinical management of breast cancer patients, especially during staging; detection of recurrence and evaluation of treatment response; the role of 16α-18Ffluoro-17β-oestradiol positron emission tomography in oestrogen receptors positive breast cancer; the promising radiopharmaceuticals, such as 89Zr-trastuzumab and 68Ga- or 18F-labeled fibroblast activation protein inhibitor; and the application of artificial intelligence.
Collapse
Affiliation(s)
- Jules Zhang-Yin
- Department of Nuclear Medicine, Clinique Sud Luxembourg, Vivalia, B-6700 Arlon, Belgium
| |
Collapse
|
19
|
Mankoff DA, Clark AS, Edmonds CE, O'Brien SR, Pantel AR. 16α-[ 18F]Fluoro-17β-Estradiol Positron Emission Tomography to Measure Regional Estrogen Receptor Expression in Breast Cancer. J Clin Oncol 2022; 40:3660-3663. [PMID: 36041088 DOI: 10.1200/jco.22.01055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- David A Mankoff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Breast Cancer Research Program, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Amy S Clark
- Breast Cancer Research Program, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Christine E Edmonds
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Sophia R O'Brien
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Austin R Pantel
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
20
|
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
|
21
|
Kurth J, Potratz M, Heuschkel M, Krause BJ, Schwarzenböck SM. GRPr Theranostics: Current Status of Imaging and Therapy using GRPr Targeting Radiopharmaceuticals. Nuklearmedizin 2022; 61:247-261. [PMID: 35668669 DOI: 10.1055/a-1759-4189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Addressing molecular targets, that are overexpressed by various tumor entities, using radiolabeled molecules for a combined diagnostic and therapeutic (theranostic) approach is of increasing interest in oncology. The gastrin-releasing peptide receptor (GRPr), which is part of the bombesin family, has shown to be overexpressed in a variety of tumors, therefore, serving as a promising target for those theranostic applications. A large amount of differently radiolabeled bombesin derivatives addressing the GRPr have been evaluated in the preclinical as well as clinical setting showing fast blood clearance and urinary excretion with selective GRPr-binding. Most of the available studies on GRPr-targeted imaging and therapy have evaluated the theranostic approach in prostate and breast cancer applying bombesin derivatives tagged with the predominantly used theranostic pair of 68Ga/177Lu which is the focus of this review.
Collapse
Affiliation(s)
- Jens Kurth
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | - Madlin Potratz
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | - Martin Heuschkel
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | - Bernd J Krause
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | | |
Collapse
|
22
|
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
|
23
|
Altunay B, Morgenroth A, Mottaghy FM. Use of Radionuclide-Based Imaging Methods in Breast Cancer. Semin Nucl Med 2022; 52:561-573. [PMID: 35624034 DOI: 10.1053/j.semnuclmed.2022.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/07/2022] [Accepted: 04/13/2022] [Indexed: 12/21/2022]
Abstract
Breast cancer is one of the most commonly occurring cancers in women globally and is the primary cause of cancer mortality in females. Thus, early and effective breast cancer diagnosis is crucial for enhancing the survival rate. Current standard diagnostic techniques to assess the hormone receptor status in biopsies include immunohistochemistry and fluorescence in situ hybridization. However, in recent years, there has been an increase in research on noninvasive techniques for molecular imaging of hormone receptors. These methods offer many advantages over conventional imaging, as repeated measurements can be used to capture heterogeneous tumor expression throughout the body, as well as transformations in receptor status during disease progression. Thus, the noninvasive method, as an adjunct to conventional imaging, offers the potential to improve patient selection, optimize dose and schedule, and streamline the assessment of response.
Collapse
Affiliation(s)
- Betül Altunay
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Agnieszka Morgenroth
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, Aachen, Germany; Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, Germany; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands.
| |
Collapse
|
24
|
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
|
25
|
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]
|
26
|
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]
|
27
|
Hatazawa J. The Clinical Value of Breast Specific Gamma Imaging and Positron Imaging: An Update. Semin Nucl Med 2022; 52:619-627. [PMID: 35346487 DOI: 10.1053/j.semnuclmed.2022.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 01/15/2023]
Abstract
In the management of patients with breast cancer (BC), a mammography contributed to screen an early-stage patient, to plan a therapy strategy, to evaluate a therapy outcome, to detect a recurrence, and to reduce a mortality. Currently, various imaging modalities, such as CT, MR, Ultrasound (US), SPECT/CT, PET/CT, PET/MR have been utilized for the management of BC patients. In order to overcome a limited spatial resolution and sensitivity of whole-body systems in nuclear medicine imaging, dedicated breast imaging modalities were developed. One is a gamma imaging system with single/dual head scintillation detectors or semiconductor detectors associated with light compression device for breast parenchyma. Radiopharmaceutical for the gamma imaging is 99mTc-sestamibi. Another is a positron imaging system with opposite-type panel detectors and ring-shaped type detectors. Radiopharmaceutical for positron imaging is 18F-fluorodeoxyglucose. The breast-specific gamma and positron imaging systems were utilized mainly to detect small lesions less than 1 cm in diameter especially in patients with dense breast, to evaluate an effect of preoperative neo-adjuvant therapy, to plan surgical procedures (conservative-surgery vs mastectomy), and to detect a recurrence. By combining higher sensitivity and spatial resolution scanners with new radiopharmaceuticals, an information on molecular-level pathology of BC is increasingly available in an individual patient. This article reviewed clinical impact and future perspective of this field.
Collapse
Affiliation(s)
- Jun Hatazawa
- Department of Quantum Cancer Therapy, Research Center for Nuclear Physics, Osaka University, Osaka, Japan; Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, Osaka, Japan; Institute for Radiation Sciences, Osaka University, Osaka, Japan; Immunology Frontier Research Center, Osaka University, Osaka, Japan.
| |
Collapse
|
28
|
Pantel AR, Viswanath V, Muzi M, Doot RK, Mankoff DA. Principles of Tracer Kinetic Analysis in Oncology, Part I: Principles and Overview of Methodology. J Nucl Med 2022; 63:342-352. [PMID: 35232879 DOI: 10.2967/jnumed.121.263518] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
Learning Objectives: On successful completion of this activity, participants should be able to describe (1) describe principles of PET tracer kinetic analysis for oncologic applications; (2) list methods used for PET kinetic analysis for oncology; and (3) discuss application of kinetic modeling for cancer-specific diagnostic needs.Financial Disclosure: This work was supported by KL2 TR001879, R01 CA211337, R01 CA113941, R33 CA225310, Komen SAC130060, R50 CA211270, and K01 DA040023. Dr. Pantel is a consultant or advisor for Progenics and Blue Earth Diagnostics and is a meeting participant or lecturer for Blue Earth Diagnostics. Dr. Mankoff is on the scientific advisory boards of GE Healthcare, Philips Healthcare, Reflexion, and ImaginAb and is the owner of Trevarx; his wife is the chief executive officer of Trevarx. The authors of this article have indicated no other relevant relationships that could be perceived as a real or apparent conflict of interest.CME Credit: SNMMI is accredited by the Accreditation Council for Continuing Medical Education (ACCME) to sponsor continuing education for physicians. SNMMI designates each JNM continuing education article for a maximum of 2.0 AMA PRA Category 1 Credits. Physicians should claim only credit commensurate with the extent of their participation in the activity. For CE credit, SAM, and other credit types, participants can access this activity through the SNMMI website (http://www.snmmilearningcenter.org) through March 2025PET enables noninvasive imaging of regional in vivo cancer biology. By engineering a radiotracer to target specific biologic processes of relevance to cancer (e.g., cancer metabolism, blood flow, proliferation, and tumor receptor expression or ligand binding), PET can detect cancer spread, characterize the cancer phenotype, and assess its response to treatment. For example, imaging of glucose metabolism using the radiolabeled glucose analog 18F-FDG has widespread applications to all 3 of these tasks and plays an important role in cancer care. However, the current clinical practice of imaging at a single time point remote from tracer injection (i.e., static imaging) does not use all the information that PET cancer imaging can provide, especially to address questions beyond cancer detection. Reliance on tracer measures obtained only from static imaging may also lead to misleading results. In this 2-part continuing education paper, we describe the principles of tracer kinetic analysis for oncologic PET (part 1), followed by examples of specific implementations of kinetic analysis for cancer PET imaging that highlight the added benefits over static imaging (part 2). This review is designed to introduce nuclear medicine clinicians to basic concepts of kinetic analysis in oncologic imaging, with a goal of illustrating how kinetic analysis can augment our understanding of in vivo cancer biology, improve our approach to clinical decision making, and guide the interpretation of quantitative measures derived from static images.
Collapse
Affiliation(s)
- Austin R Pantel
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Varsha Viswanath
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Mark Muzi
- Department of Radiology, University of Washington, Seattle, Washington
| | - Robert K Doot
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - David A Mankoff
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania; and
| |
Collapse
|
29
|
Franquet E, Park H. Molecular imaging in oncology: Common PET/CT radiopharmaceuticals and applications. Eur J Radiol Open 2022; 9:100455. [DOI: 10.1016/j.ejro.2022.100455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/26/2022] Open
|
30
|
PET imaging in breast cancer. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00124-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
31
|
Parihar AS, Bhattacharya A. Role of Nuclear Medicine in Breast Cancer. Breast Cancer 2022. [DOI: 10.1007/978-981-16-4546-4_12] [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]
|
32
|
Young AJ, Pantel AR, Viswanath V, Dominguez TL, Makvandi M, Lee H, Li S, Schubert EK, Pryma DA, Farwell MD, Mach RH, Simpkins F, Lin LL, Mankoff DA, Doot RK. Kinetic and Static Analysis of Poly-(Adenosine Diphosphate-Ribose) Polymerase-1-Targeted 18F-Fluorthanatrace PET Images of Ovarian Cancer. J Nucl Med 2022; 63:44-50. [PMID: 33863820 PMCID: PMC8717190 DOI: 10.2967/jnumed.121.261894] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 03/26/2021] [Indexed: 12/21/2022] Open
Abstract
The poly-(adenosine diphosphate-ribose) polymerase (PARP) family of proteins participates in numerous functions, most notably the DNA damage response. Cancer vulnerability to DNA damage has led to development of several PARP inhibitors (PARPi). This class of drugs has demonstrated therapeutic efficacy in ovarian, breast, and prostate cancers, but with variable response. Consequently, clinics need to select patients likely to benefit from these targeted therapies. In vivo imaging of 18F-fluorthanatrace uptake has been shown to correspond to PARP-1 expression in tissue. This study characterized the pharmacokinetics of 18F-fluorthanatrace and tested kinetic and static models to guide metric selection in future studies assessing 18F-fluorthanatrace as a biomarker of response to PARPi therapy. Methods: Fourteen prospectively enrolled ovarian cancer patients were injected with 18F-fluorthanatrace and imaged dynamically for 60 min after injection followed by up to 2 whole-body scans, with venous blood activity and metabolite measurements. SUVmax and SUVpeak were extracted from dynamic images and whole-body scans. Kinetic parameter estimates and SUVs were assessed for correlations with tissue PARP-1 immunofluorescence (n = 7). Simulations of population kinetic parameters enabled estimation of measurement bias and precision in parameter estimates. Results:18F-fluorthanatrace blood clearance was variable, but labeled metabolite profiles were similar across patients, supporting use of a population parent fraction curve. The total distribution volume from a reversible 2-tissue-compartment model and Logan reference tissue distribution volume ratio (DVR) from the first hour of PET acquisition correlated with tumor PARP-1 expression by immunofluorescence (r = 0.76 and 0.83, respectively; P < 0.05). DVR bias and precision estimates were 6.4% and 29.1%, respectively. SUVmax and SUVpeak acquired from images with midpoints of 57.5, 110 ± 3, and 199 ± 4 min highly correlated with PARP-1 expression (mean ± SD, r ≥ 0.79; P < 0.05). Conclusion: Tumor SUVmax and SUVpeak at 55-60 min after injection and later and DVR from at least 60 min appear to be robust noninvasive measures of PARP-1 binding. 18F-fluorthanatrace uptake in ovarian cancer was best described by models of reversible binding. However, pharmacokinetic patterns of tracer uptake were somewhat variable, especially at later time points.
Collapse
Affiliation(s)
- Anthony J Young
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Austin R Pantel
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Varsha Viswanath
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Tiffany L Dominguez
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mehran Makvandi
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hsiaoju Lee
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shihong Li
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Erin K Schubert
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniel A Pryma
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael D Farwell
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert H Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Fiona Simpkins
- Division of Gynecology and Oncology, Department of OBGYN, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Lilie L Lin
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David A Mankoff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert K Doot
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania;
| |
Collapse
|
33
|
Yu Y, Liu A, Dhawan G, Mei H, Zhang W, Izawa K, Soloshonok VA, Han J. Fluorine-containing pharmaceuticals approved by the FDA in 2020: Synthesis and biological activity. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2021.05.042] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
34
|
Takahashi M, Maeda H, Tsujikawa T, Kono H, Mori T, Kiyono Y, Okazawa H, Noriki S, Imamura Y, Goi T. 18F-Fluoroestradiol Tumor Uptake Is Influenced by Structural Components in Breast Cancer. Clin Nucl Med 2021; 46:884-889. [PMID: 34319950 DOI: 10.1097/rlu.0000000000003835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE Estrogen receptor (ER) is expressed in the majority of invasive breast cancer and is an important prognostic indicator. The tumor stroma also plays an important role in disease progression. This study evaluated the effect of stromal components on 16α-[18F]-fluoro-17β-estradiol (18F-FES) uptake in breast cancer and proposed a partial-volume correction method for 18F-FES PET based on histopathological analyses. PATIENTS AND METHODS Fifteen patients with biopsy-confirmed breast cancer underwent preoperative 18F-FES PET. Estrogen receptor expression in biopsy specimens was assayed by immunohistochemistry, cellular components in surgical specimens were measured using hematoxylin-eosin staining, and nuclear components in surgical and biopsy specimens were measured using Azan-Mallory staining. The relationship between 18F-FES SUV of the primary tumor and histopathological findings including ER expression, the Allred score, ER-positive cellular component ratio, and ER-positive nuclear component ratio (NCR) was examined. The relationship between stroma-free 18F-FES SUV and ER expression was also examined. RESULTS 18F-FES uptake was not significantly positively correlated with ER expression (r = 0.44, P = 0.10). 18F-FES uptake was significantly correlated with the Allred score, ER-positive cellular component ratio, and ER-positive NCR in surgical specimens (ρ = 0.60, P = 0.02; r = 0.55, P = 0.03; and r = 0.65, P = 0.01, respectively). 18F-FES uptake was predominantly correlated with ER-positive NCR in biopsy specimens (r = 0.84, P < 0.001). Stroma-free 18F-FES SUV was significantly correlated with ER expression (r = 0.78, P < 0.01). CONCLUSIONS 18F-FES PET predominantly demonstrates the level of ER expression in breast cancer cell nucleus. Although tumor 18F-FES uptake is affected by the degree of stromal components, the partial volume effect on the uptake can be corrected by stroma-volume fraction in Azan-Mallory staining.
Collapse
Affiliation(s)
- Mizuho Takahashi
- From the First Department of Surgery, Faculty of Medical Sciences
| | - Hiroyuki Maeda
- From the First Department of Surgery, Faculty of Medical Sciences
| | | | - Hiroko Kono
- From the First Department of Surgery, Faculty of Medical Sciences
| | - Tetsuya Mori
- Biomedical Imaging Research Center, University of Fukui
| | | | | | - Sakon Noriki
- Faculty of Nursing and Social Welfare Science, Fukui Prefectural University
| | - Yoshiaki Imamura
- Division of Surgical Pathology, University of Fukui Hospital, Fukui, Japan
| | - Takanori Goi
- From the First Department of Surgery, Faculty of Medical Sciences
| |
Collapse
|
35
|
Radionuclide-Based Imaging of Breast Cancer: State of the Art. Cancers (Basel) 2021; 13:cancers13215459. [PMID: 34771622 PMCID: PMC8582396 DOI: 10.3390/cancers13215459] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/14/2021] [Accepted: 10/14/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Breast cancer is one of the most commonly diagnosed malignant tumors, possessing high incidence and mortality rates that threaten women’s health. Thus, early and effective breast cancer diagnosis is crucial for enhancing the survival rate. Radionuclide molecular imaging displays its advantages for detecting breast cancer from a functional perspective. Noninvasive visualization of biological processes with radionuclide-labeled small metabolic compounds helps elucidate the metabolic state of breast cancer, while radionuclide-labeled ligands/antibodies for receptor-targeted radionuclide molecular imaging is sensitive and specific for visualization of the overexpressed molecular markers in breast cancer. This review focuses on the most recent developments of novel radiotracers as promising tools for early breast cancer diagnosis. Abstract Breast cancer is a malignant tumor that can affect women worldwide and endanger their health and wellbeing. Early detection of breast cancer can significantly improve the prognosis and survival rate of patients, but with traditional anatomical imagine methods, it is difficult to detect lesions before morphological changes occur. Radionuclide-based molecular imaging based on positron emission tomography (PET) and single-photon emission computed tomography (SPECT) displays its advantages for detecting breast cancer from a functional perspective. Radionuclide labeling of small metabolic compounds can be used for imaging biological processes, while radionuclide labeling of ligands/antibodies can be used for imaging receptors. Noninvasive visualization of biological processes helps elucidate the metabolic state of breast cancer, while receptor-targeted radionuclide molecular imaging is sensitive and specific for visualization of the overexpressed molecular markers in breast cancer, contributing to early diagnosis and better management of cancer patients. The rapid development of radionuclide probes aids the diagnosis of breast cancer in various aspects. These probes target metabolism, amino acid transporters, cell proliferation, hypoxia, estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), gastrin-releasing peptide receptor (GRPR) and so on. This article provides an overview of the development of radionuclide molecular imaging techniques present in preclinical or clinical studies, which are used as tools for early breast cancer diagnosis.
Collapse
|
36
|
Wawruszak A, Borkiewicz L, Okon E, Kukula-Koch W, Afshan S, Halasa M. Vorinostat (SAHA) and Breast Cancer: An Overview. Cancers (Basel) 2021; 13:4700. [PMID: 34572928 PMCID: PMC8468501 DOI: 10.3390/cancers13184700] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/12/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023] Open
Abstract
Vorinostat (SAHA), an inhibitor of class I and II of histone deacetylases, is the first histone deacetylase inhibitor (HDI) approved for the treatment of cutaneous T-cell lymphoma in 2006. HDIs are promising anticancer agents that inhibit the proliferation of many types of cancer cells including breast carcinoma (BC). BC is a heterogeneous disease with variable biological behavior, morphological features, and response to therapy. Although significant progress in the treatment of BC has been made, high toxicity to normal cells, serious side effects, and the occurrence of multi-drug resistance limit the effective therapy of BC patients. Therefore, new active agents which improve the effectiveness of currently used regimens are highly needed. This manuscript analyzes preclinical and clinical trials data of SAHA, applied individually or in combination with other anticancer agents, considering different histological subtypes of BC.
Collapse
Affiliation(s)
- Anna Wawruszak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (L.B.); (E.O.); (M.H.)
| | - Lidia Borkiewicz
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (L.B.); (E.O.); (M.H.)
| | - Estera Okon
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (L.B.); (E.O.); (M.H.)
| | - Wirginia Kukula-Koch
- Department of Pharmacognosy, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Syeda Afshan
- Institute of Biomedicine and FICAN West Cancer Centre, University of Turku, 20521 Turku, Finland;
| | - Marta Halasa
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (L.B.); (E.O.); (M.H.)
| |
Collapse
|
37
|
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
|
38
|
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]
|
39
|
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
|
40
|
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
|
41
|
Automated synthesis of the 16α-[18F]fluoroestradiol ([18F]FES): minimization of precursor amount and resulting benefits. RADIOCHIM ACTA 2020. [DOI: 10.1515/ract-2020-0058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Abstract
The 16α-[18F]Fluoroestradiol ([18F]FES) is an established PET radiotracer for estrogen positive (ER+) breast cancer. Although the radiosynthesis is well-described, the majority of the published methods suffer from modest or irreproducible yields and time-intensive purification procedures. In view of the considerable clinical applications, development of a more efficient and faster synthesis of [18F]FES still remains a task of a significant practical importance. [18F]FES was produced by a direct nucleophilic radiofluorination of 3-O-methoxymethyl-16,17-O-sulfuryl-16-epiestriol (MMSE), followed by acidic hydrolysis using HCl/CH3CN. [18F]Fluoride retained on a QMA carb cartridge (46 mg) was eluted by solution of 1.2 mg of tetrabutylammonium tosylate (TBAOTs) in EtOH. After fluorination reaction (0.3 mg MMSE, 1 ml of CH3CN/100 °C, 5 min) [18F]FES was isolated by single-cartridge SPE purification using OASIS WAX 3cc, elution accomplished with aqueous ethanol of different concentrations. On а GE TRACERlab FX N Pro automated module [18F]FES (formulated in normal saline with 5% EtOH) was obtained in 33 ± 3% yield (n = 5, non-decay corrected) within 32 min. Reduction of precursor amount, exclusion of azeotropic drying step and simplification of purification make the suggested method readily adaptable to various automated synthesizers and offers significant cost decrease.
Collapse
|
42
|
Gillman JA, Pantel AR, Mankoff DA, Edmonds CE. Update on Quantitative Imaging for Predicting and Assessing Response in Oncology. Semin Nucl Med 2020; 50:505-517. [PMID: 33059820 PMCID: PMC9788668 DOI: 10.1053/j.semnuclmed.2020.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Molecular imaging has revolutionized clinical oncology by imaging-specific facets of cancer biology. Through noninvasive measurements of tumor physiology, targeted radiotracers can serve as biomarkers for disease characterization, prognosis, response assessment, and predicting long-term response/survival. In turn, these imaging biomarkers can be utilized to tailor therapeutic regimens to tumor biology. In this article, we review biomarker applications for response assessment and predicting long-term outcomes. 18F-fluorodeoxyglucose (FDG), a measure of cellular glucose metabolism, is discussed in the context of lymphoma and breast and lung cancer. FDG has gained widespread clinical acceptance and has been integrated into the routine clinical care of several malignancies, most notably lymphoma. The novel radiotracers 16α-18F-fluoro-17β-estradiol and 18F-fluorothymidine are reviewed in application to the early prediction of response assessment of breast cancer. Through illustrative examples, we explore current and future applications of molecular imaging biomarkers in the advancement of precision medicine.
Collapse
Affiliation(s)
- Jennifer A Gillman
- Department of Radiology, Division of Nuclear Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Austin R Pantel
- Department of Radiology, Division of Nuclear Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - David A Mankoff
- Department of Radiology, Division of Nuclear Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Christine E Edmonds
- Department of Radiology, Division of Nuclear Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA.
| |
Collapse
|
43
|
Nimmagadda S. Quantifying PD-L1 Expression to Monitor Immune Checkpoint Therapy: Opportunities and Challenges. Cancers (Basel) 2020; 12:cancers12113173. [PMID: 33137949 PMCID: PMC7692040 DOI: 10.3390/cancers12113173] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 10/19/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Malignant cells hijack the regulatory roles of immune checkpoint proteins for immune evasion and survival. Therapeutics blocking those proteins can restore the balance of the immune system and lead to durable responses in cancer patients. Although a subset of patients derive benefit, there are few non-invasive technologies to guide and monitor those therapies to improve success rates. This is a review of the advancements in non-invasive methods for quantification of immune checkpoint protein programmed death ligand 1 expression, a biomarker detected by immunohistochemistry and widely used for guiding immune checkpoint therapy. Abstract Therapeutics targeting programmed death ligand 1 (PD-L1) protein and its receptor PD-1 are now dominant players in restoring anti-tumor immune responses. PD-L1 detection by immunohistochemistry (IHC) is emerging as a reproducible biomarker for guiding patient stratification for those therapies in some cancers. However, PD-L1 expression in the tumor microenvironment is highly complex. It is upregulated by aberrant genetic alterations, and is highly regulated at the transcriptional, posttranscriptional, and protein levels. Thus, PD-L1 IHC is inadequate to fully understand the relevance of PD-L1 levels in the whole body and their dynamics to improve therapeutic outcomes. Imaging technologies could potentially assist in meeting that need. Early clinical investigations show promising results in quantifying PD-L1 expression in the whole body by positron emission tomography (PET). Within this context, this review summarizes advancements in regulation of PD-L1 expression and imaging agents, and in PD-L1 PET for drug development, and discusses opportunities and challenges presented by these innovations for guiding immune checkpoint therapy (ICT).
Collapse
Affiliation(s)
- Sridhar Nimmagadda
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; ; Tel.: +1-410-502-6244; Fax: +1-410-614-3147
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Bloomberg–Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
44
|
Heesch A, Maurer J, Stickeler E, Beheshti M, Mottaghy FM, Morgenroth A. Development of Radiotracers for Breast Cancer-The Tumor Microenvironment as an Emerging Target. Cells 2020; 9:cells9102334. [PMID: 33096754 PMCID: PMC7590199 DOI: 10.3390/cells9102334] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/19/2020] [Accepted: 10/19/2020] [Indexed: 12/17/2022] Open
Abstract
Molecular imaging plays an increasingly important role in the diagnosis and treatment of different malignancies. Radiolabeled probes enable the visualization of the primary tumor as well as the metastases and have been also employed in targeted therapy and theranostic approaches. With breast cancer being the most common malignancy in women worldwide it is of special interest to develop novel targeted treatments. However, tumor microenvironment and escape mechanisms often limit their therapeutic potential. Addressing tumor stroma associated targets provides a promising option to inhibit tumor growth and angiogenesis and to disrupt tumor tissue architecture. This review describes recent developments on radiolabeled probes used in diagnosis and treatment of breast cancer especially in triple negative type with the focus on potential targets offered by the tumor microenvironment, like tumor associated macrophages, cancer associated fibroblasts, and endothelial cells.
Collapse
Affiliation(s)
- Amelie Heesch
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany; (A.H.); (M.B.); (F.M.M.)
| | - Jochen Maurer
- Department of Obstetrics and Gynecology, University Hospital Aachen (UKA), 52074 Aachen, Germany; (J.M.); (E.S.)
| | - Elmar Stickeler
- Department of Obstetrics and Gynecology, University Hospital Aachen (UKA), 52074 Aachen, Germany; (J.M.); (E.S.)
| | - Mohsen Beheshti
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany; (A.H.); (M.B.); (F.M.M.)
- Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Felix M. Mottaghy
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany; (A.H.); (M.B.); (F.M.M.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), 6202 Maastricht, The Netherlands
| | - Agnieszka Morgenroth
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany; (A.H.); (M.B.); (F.M.M.)
- Correspondence:
| |
Collapse
|
45
|
Kurland BF, Wiggins JR, Coche A, Fontan C, Bouvet Y, Webner P, Divgi C, Linden HM. Whole-Body Characterization of Estrogen Receptor Status in Metastatic Breast Cancer with 16α-18F-Fluoro-17β-Estradiol Positron Emission Tomography: Meta-Analysis and Recommendations for Integration into Clinical Applications. Oncologist 2020; 25:835-844. [PMID: 32374053 PMCID: PMC7543360 DOI: 10.1634/theoncologist.2019-0967] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/02/2020] [Indexed: 12/19/2022] Open
Abstract
Estrogen receptor (ER) status by immunohistochemistry (IHC) of cancer tissue is currently used to direct endocrine therapy in breast cancer. Positron emission tomography (PET) with 16α-18F-fluoro-17β-estradiol (18 F-FES) noninvasively characterizes ER ligand-binding function of breast cancer lesions. Concordance of imaging and tissue assays should be established for 18 F-FES PET to be an alternative or complement to tissue biopsy for metastatic lesions. We conducted a meta-analysis of published results comparing 18 F-FES PET and tissue assays of ER status in patients with breast cancer. PubMed and EMBASE were searched for English-language manuscripts with at least 10 patients and low overall risk of bias. Thresholds for imaging and tissue classification could differ between studies but had to be clearly stated. We used hierarchical summary receiver-operating characteristic curve models for the meta-analysis. The primary analysis included 113 nonbreast lesions from 4 studies; an expanded analysis included 327 total lesions from 11 studies. Treating IHC results as the reference standard, sensitivity was 0.78 (95% confidence region 0.65-0.88) and specificity 0.98 (0.65-1.00) for the primary analysis of nonbreast lesions. In the expanded analysis including non-IHC tissue assays and all lesion sites, sensitivity was 0.81 (0.73-0.87) and specificity 0.86 (0.68-0.94). These results suggest that 18 F-FES PET is useful for characterization of ER status of metastatic breast cancer lesions. We also review current best practices for conducting 18 F-FES PET scans. This imaging assay has potential to improve clinically relevant outcomes for patients with (historically) ER-positive metastatic breast cancer, including those with brain metastases and/or lobular histology. IMPLICATIONS FOR PRACTICE: 16α-18F-fluoro-17β-estradiol positron emission tomography (18 F-FES PET) imaging assesses estrogen receptor status in breast cancer in vivo. This work reviews the sensitivity and specificity of 18 F-FES PET in a meta-analysis with reference tissue assays and discusses best practices for use of the tracer as an imaging biomarker. 18 F-FES PET could enhance breast cancer diagnosis and staging as well as aid in therapy selection for patients with metastatic disease. Tissue sampling limitations, intrapatient heterogeneity, and temporal changes in molecular markers make it likely that 18 F-FES PET will complement existing assays when clinically available in the near future.
Collapse
Affiliation(s)
| | - Jay R. Wiggins
- Merlin Biomedical Consulting, LLCHendersonvilleNorth CarolinaUSA
| | | | | | | | | | | | | |
Collapse
|
46
|
Ming Y, Wu N, Qian T, Li X, Wan DQ, Li C, Li Y, Wu Z, Wang X, Liu J, Wu N. Progress and Future Trends in PET/CT and PET/MRI Molecular Imaging Approaches for Breast Cancer. Front Oncol 2020; 10:1301. [PMID: 32903496 PMCID: PMC7435066 DOI: 10.3389/fonc.2020.01301] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 06/23/2020] [Indexed: 12/13/2022] Open
Abstract
Breast cancer is a major disease with high morbidity and mortality in women worldwide. Increased use of imaging biomarkers has been shown to add more information with clinical utility in the detection and evaluation of breast cancer. To date, numerous studies related to PET-based imaging in breast cancer have been published. Here, we review available studies on the clinical utility of different PET-based molecular imaging methods in breast cancer diagnosis, staging, distant-metastasis detection, therapeutic and prognostic prediction, and evaluation of therapeutic responses. For primary breast cancer, PET/MRI performed similarly to MRI but better than PET/CT. PET/CT and PET/MRI both have higher sensitivity than MRI in the detection of axillary and extra-axillary nodal metastases. For distant metastases, PET/CT has better performance in the detection of lung metastasis, while PET/MRI performs better in the liver and bone. Additionally, PET/CT is superior in terms of monitoring local recurrence. The progress in novel radiotracers and PET radiomics presents opportunities to reclassify tumors by combining their fine anatomical features with molecular characteristics and develop a beneficial pathway from bench to bedside to predict the treatment response and prognosis of breast cancer. However, further investigation is still needed before application of these modalities in clinical practice. In conclusion, PET-based imaging is not suitable for early-stage breast cancer, but it adds value in identifying regional nodal disease and distant metastases as an adjuvant to standard diagnostic imaging. Recent advances in imaging techniques would further widen the comprehensive and convergent applications of PET approaches in the clinical management of breast cancer.
Collapse
Affiliation(s)
- Yue Ming
- PET-CT Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Wu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, China
| | - Tianyi Qian
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Li
- Department of Radiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - David Q Wan
- Department of Diagnostic and Interventional Imaging, McGovern Medical School, Health and Science Center at Houston, University of Texas, Houston, TX, United States
| | - Caiying Li
- Department of Medical Imaging, Second Hospital of Hebei Medical University, Hebei, China
| | - Yalun Li
- Department of Breast Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Zhihong Wu
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, China.,Department of Central Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiang Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaqi Liu
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ning Wu
- PET-CT Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
47
|
Seenu V, Sharma A, Kumar R, Suhani S, Prashanth A, Mathur S, Parshad R. Evaluation of estrogen expression of breast cancer using 18F-FES PET CT-A novel technique. World J Nucl Med 2020; 19:233-239. [PMID: 33354178 PMCID: PMC7745867 DOI: 10.4103/wjnm.wjnm_71_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 01/13/2020] [Indexed: 11/20/2022] Open
Abstract
Estrogen receptor (ER) expression in breast cancer is routinely studied on immunohistochemistry (IHC) of tissue obtained from core biopsy or surgical specimen. Sampling error and heterogeneity of tumor may incorrectly label a breast tumor as ER negative, thus denying patient hormonal treatment. Molecular functional ER imaging can assess the in-vivo ER expression of primary tumor and metastases at sites inaccessible for biopsy and also track changes in expression over time. The aim was to study ER expression using 16α-18F-fluoro-17β-estradiol or 18F-fluoroestradiol (18F FES) positron emission tomography (PET) computed tomography (CT). Twenty-four biopsy-proven breast cancer patients consenting to participate in the study underwent FES PET CT. Standard uptake value (SUVmean) of maximum of 7 lesions/patient was analyzed, and tumor-to-background ratio was calculated for each lesion. Visual interpretation score was calculated for lesion on FES PET and correlated with the Allred score on IHC of tumor tissue samples for ER expression. The diagnostic indices of FES PET CT were assessed taking IHC as “gold standard.” On FES PET CT, the mean SUV for ER+ tumors was 4.75, whereas the mean SUV for ER − tumors was 1.41. Using receiver operating characteristic curve, tumors with an SUV of ≥ 1.8 on FES PET could be considered as ER+. The overall accuracy of FES PET CT to detect ER expression was 91.66%, with two false negatives noted in this study. 18F-FES PET CT appears promising in evaluating ER expression in breast cancer. It is noninvasive and has potential to assess the in-vivo ER expression of the entire primary tumor and metastasis not amenable for biopsy.
Collapse
Affiliation(s)
- Vuthaluru Seenu
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, India
| | - Ankit Sharma
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, India
| | - Rakesh Kumar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Suhani Suhani
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, India
| | - Arun Prashanth
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Sandeep Mathur
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Rajinder Parshad
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
48
|
Peterson LM, Kurland BF, Yan F, Jiresova AN, Gadi VK, Specht JM, Gralow JR, Schubert EK, Link JM, Krohn KA, Eary JF, Mankoff DA, Linden HM. 18F-Fluoroestradiol PET Imaging in a Phase II Trial of Vorinostat to Restore Endocrine Sensitivity in ER+/HER2- Metastatic Breast Cancer. J Nucl Med 2020; 62:184-190. [PMID: 32591490 DOI: 10.2967/jnumed.120.244459] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/27/2020] [Indexed: 12/23/2022] Open
Abstract
Histone deacetylase inhibitors (HDACIs) may overcome endocrine resistance in estrogen receptor-positive (ER+) metastatic breast cancer. We tested whether 18F-fluoroestradiol PET imaging would elucidate the pharmacodynamics of combination HDACIs and endocrine therapy. Methods: Patients with ER+/human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer with prior clinical benefit from endocrine therapy but later progression on aromatase inhibitor (AI) therapy were given vorinostat (400 mg daily) sequentially or simultaneously with AI. 18F-fluoroestradiol PET and 18F-FDG PET scans were performed at baseline, week 2, and week 8. Results: Eight patients were treated sequentially, and then 15 simultaneously. Eight patients had stable disease at week 8, and 6 of these 8 patients had more than 6 mo of stable disease. Higher baseline 18F-fluoroestradiol uptake was associated with longer progression-free survival. 18F-fluoroestradiol uptake did not systematically increase with vorinostat exposure, indicating no change in regional ER estradiol binding, and 18F-FDG uptake did not show a significant decrease, as would have been expected with tumor regression. Conclusion: Simultaneous HDACIs and AI dosing in patients with cancer resistant to AI alone showed clinical benefit (6 or more months without progression) in 4 of 10 evaluable patients. Higher 18F-fluoroestradiol PET uptake identified patients likely to benefit from combination therapy, but vorinostat did not change ER expression at the level of detection of 18F-fluoroestradiol PET.
Collapse
Affiliation(s)
- Lanell M Peterson
- Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, Seattle, Washington
| | - Brenda F Kurland
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Fengting Yan
- Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, Seattle, Washington
| | - Alena Novakova- Jiresova
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Vijayakrishna K Gadi
- Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, Seattle, Washington.,Clinical Research and Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Jennifer M Specht
- Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, Seattle, Washington
| | - Julie R Gralow
- Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, Seattle, Washington
| | - Erin K Schubert
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jeanne M Link
- Department of Diagnostic Radiology, Oregon Health and Science University, Portland, Oregon; and
| | - Kenneth A Krohn
- Department of Diagnostic Radiology, Oregon Health and Science University, Portland, Oregon; and
| | - Janet F Eary
- Cancer Imaging Program, National Cancer Institute, Bethesda, Maryland
| | - David A Mankoff
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hannah M Linden
- Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, Seattle, Washington
| |
Collapse
|
49
|
PET/MRI in breast cancer patients: Added value, barriers to implementation, and solutions. Clin Imaging 2020; 68:24-28. [PMID: 32562923 DOI: 10.1016/j.clinimag.2020.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/18/2020] [Accepted: 06/01/2020] [Indexed: 11/21/2022]
|
50
|
Jones EF, Hathi DK, Freimanis R, Mukhtar RA, Chien AJ, Esserman LJ, van’t Veer LJ, Joe BN, Hylton NM. Current Landscape of Breast Cancer Imaging and Potential Quantitative Imaging Markers of Response in ER-Positive Breast Cancers Treated with Neoadjuvant Therapy. Cancers (Basel) 2020; 12:E1511. [PMID: 32527022 PMCID: PMC7352259 DOI: 10.3390/cancers12061511] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 12/24/2022] Open
Abstract
In recent years, neoadjuvant treatment trials have shown that breast cancer subtypes identified on the basis of genomic and/or molecular signatures exhibit different response rates and recurrence outcomes, with the implication that subtype-specific treatment approaches are needed. Estrogen receptor-positive (ER+) breast cancers present a unique set of challenges for determining optimal neoadjuvant treatment approaches. There is increased recognition that not all ER+ breast cancers benefit from chemotherapy, and that there may be a subset of ER+ breast cancers that can be treated effectively using endocrine therapies alone. With this uncertainty, there is a need to improve the assessment and to optimize the treatment of ER+ breast cancers. While pathology-based markers offer a snapshot of tumor response to neoadjuvant therapy, non-invasive imaging of the ER disease in response to treatment would provide broader insights into tumor heterogeneity, ER biology, and the timing of surrogate endpoint measurements. In this review, we provide an overview of the current landscape of breast imaging in neoadjuvant studies and highlight the technological advances in each imaging modality. We then further examine some potential imaging markers for neoadjuvant treatment response in ER+ breast cancers.
Collapse
Affiliation(s)
- Ella F. Jones
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94115, USA; (D.K.H.); (R.F.); (B.N.J.); (N.M.H.)
| | - Deep K. Hathi
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94115, USA; (D.K.H.); (R.F.); (B.N.J.); (N.M.H.)
| | - Rita Freimanis
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94115, USA; (D.K.H.); (R.F.); (B.N.J.); (N.M.H.)
| | - Rita A. Mukhtar
- Department of Surgery, University of California, San Francisco, CA 94115, USA;
| | - A. Jo Chien
- School of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA; (A.J.C.); (L.J.v.V.)
| | - Laura J. Esserman
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA;
| | - Laura J. van’t Veer
- School of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA; (A.J.C.); (L.J.v.V.)
| | - Bonnie N. Joe
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94115, USA; (D.K.H.); (R.F.); (B.N.J.); (N.M.H.)
| | - Nola M. Hylton
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94115, USA; (D.K.H.); (R.F.); (B.N.J.); (N.M.H.)
| |
Collapse
|