1
|
Ji M, Wang X, Liu C, Ma G, Lu X, Zhu B, He S, Zhang J, Xu X, Song S, Yang Z. Imaging CDK4/6 Broaden Options of Breast Cancer Diagnostics with Positron Emission Tomography. J Med Chem 2025; 68:4635-4649. [PMID: 39945599 DOI: 10.1021/acs.jmedchem.4c02672] [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: 02/28/2025]
Abstract
This study developed a novel PET radiotracer to screen breast cancer patients sensitive to CDK4/6 inhibitors, guiding personalized treatment. Two CDK4/6-targeting precursors were synthesized and evaluated in vitro and in vivo. Three breast cancer cell lines─MCF-7, MDA-MB-231, and MDA-MB-468─were selected based on decreasing sensitivity to palbociclib. Compared to [68Ga]Ga-DOTA-Hexa-CDKi, [68Ga]Ga-DOTA-Bua-CDKi clearly identified cell lines with high sensitivity to palbociclib. PET/CT imaging showed significantly higher uptake of [68Ga]Ga-DOTA-Bua-CDKi (8.40 ± 0.85%ID/g) in MCF-7 tumors 60 min after tracer injection, with significant differences in tumor uptake among the three models (P < 0.05). Blocking assays demonstrated specific tumor uptake of [68Ga]Ga-DOTA-Bua-CDKi. Biosafety tests validated its safety as a diagnostic agent. [68Ga]Ga-DOTA-Bua-CDKi showed highly specific targeting of CDK4/6 and effective contrast imaging in tumor models. To our knowledge, [68Ga]Ga-DOTA-Bua-CDKi is one of the first radiotracers to assess CDK inhibitor sensitivity, offering promise for evaluating patient responses to CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Mengjing Ji
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, China
| | - Xiangwei Wang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, China
| | - Cheng Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, China
| | - Guang Ma
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, China
| | - Xin Lu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, China
| | - Bin Zhu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, China
| | - Simin He
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, China
| | - Jianping Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, China
| | - Xiaoping Xu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, China
| | - Zhongyi Yang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, China
| |
Collapse
|
2
|
Haidar M, Rizkallah J, El Sardouk O, El Ghawi N, Omran N, Hammoud Z, Saliba N, Tfayli A, Moukadem H, Berjawi G, Nassar L, Marafi F, Choudhary P, Dadgar H, Sadeq A, Abi-Ghanem AS. Radiotracer Innovations in Breast Cancer Imaging: A Review of Recent Progress. Diagnostics (Basel) 2024; 14:1943. [PMID: 39272726 PMCID: PMC11394464 DOI: 10.3390/diagnostics14171943] [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: 07/31/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
This review focuses on the pivotal role of radiotracers in breast cancer imaging, emphasizing their importance in accurate detection, staging, and treatment monitoring. Radiotracers, labeled with radioactive isotopes, are integral to various nuclear imaging techniques, including positron emission tomography (PET) and positron emission mammography (PEM). The most widely used radiotracer in breast cancer imaging is 18F-fluorodeoxyglucose (18F-FDG), which highlights areas of increased glucose metabolism, a hallmark of many cancer cells. This allows for the identification of primary tumors and metastatic sites and the assessment of tumor response to therapy. In addition to 18F-FDG, this review will explore newer radiotracers targeting specific receptors, such as estrogen receptors or HER2, which offer more personalized imaging options. These tracers provide valuable insights into the molecular characteristics of tumors, aiding in tailored treatment strategies. By integrating radiotracers into breast cancer management, clinicians can enhance early disease detection, monitor therapeutic efficacy, and guide interventions, ultimately improving patient outcomes. Ongoing research aimed at developing more specific and sensitive tracers will also be highlighted, underscoring their potential to advance precision medicine in breast cancer care.
Collapse
Affiliation(s)
- Mohamad Haidar
- Department of Diagnostic Radiology, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Joe Rizkallah
- Department of Diagnostic Radiology, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Omar El Sardouk
- Department of Diagnostic Radiology, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Nour El Ghawi
- Department of Diagnostic Radiology, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Nadine Omran
- Department of Diagnostic Radiology, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Zeinab Hammoud
- Department of Diagnostic Radiology, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Nina Saliba
- Department of Diagnostic Radiology, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Arafat Tfayli
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Hiba Moukadem
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Ghina Berjawi
- Department of Diagnostic Radiology, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Lara Nassar
- Department of Diagnostic Radiology, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Fahad Marafi
- Jaber Al-Ahmad Centre for Molecular Imaging, Kuwait City 70031, Kuwait
| | - Partha Choudhary
- Department of Nuclear Medicine, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi 110085, India
| | - Habibollah Dadgar
- Cancer Research Center, RAZAVI Hospital, Imam Reza International University, Mashhad 9198613636, Iran
| | - Alyaa Sadeq
- Jaber Al-Ahmad Centre for Molecular Imaging, Kuwait City 70031, Kuwait
| | - Alain S Abi-Ghanem
- Department of Diagnostic Radiology, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon
| |
Collapse
|
3
|
Lu Y, Houson HA, Gallegos CA, Mascioni A, Jia F, Aivazian A, Song PN, Lynch SE, Napier TS, Mansur A, Larimer BM, Lapi SE, Hanker AB, Sorace AG. Evaluating the immunologically "cold" tumor microenvironment after treatment with immune checkpoint inhibitors utilizing PET imaging of CD4 + and CD8 + T cells in breast cancer mouse models. Breast Cancer Res 2024; 26:104. [PMID: 38918836 PMCID: PMC11201779 DOI: 10.1186/s13058-024-01844-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 05/17/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Immune-positron emission tomography (PET) imaging with tracers that target CD8 and granzyme B has shown promise in predicting the therapeutic response following immune checkpoint blockade (ICB) in immunologically "hot" tumors. However, immune dynamics in the low T-cell infiltrating "cold" tumor immune microenvironment during ICB remain poorly understood. This study uses molecular imaging to evaluate changes in CD4 + T cells and CD8 + T cells during ICB in breast cancer models and examines biomarkers of response. METHODS [89Zr]Zr-DFO-CD4 and [89Zr]Zr-DFO-CD8 radiotracers were used to quantify changes in intratumoral and splenic CD4 T cells and CD8 T cells in response to ICB treatment in 4T1 and MMTV-HER2 mouse models, which represent immunologically "cold" tumors. A correlation between PET quantification metrics and long-term anti-tumor response was observed. Further biological validation was obtained by autoradiography and immunofluorescence. RESULTS Following ICB treatment, an increase in the CD8-specific PET signal was observed within 6 days, and an increase in the CD4-specific PET signal was observed within 2 days in tumors that eventually responded to immunotherapy, while no significant differences in CD4 or CD8 were found at the baseline of treatment that differentiated responders from nonresponders. Furthermore, mice whose tumors responded to ICB had a lower CD8 PET signal in the spleen and a higher CD4 PET signal in the spleen compared to non-responders. Intratumoral spatial heterogeneity of the CD8 and CD4-specific PET signals was lower in responders compared to non-responders. Finally, PET imaging, autoradiography, and immunofluorescence signals were correlated when comparing in vivo imaging to ex vivo validations. CONCLUSIONS CD4- and CD8-specific immuno-PET imaging can be used to characterize the in vivo distribution of CD4 + and CD8 + T cells in response to immune checkpoint blockade. Imaging metrics that describe the overall levels and distribution of CD8 + T cells and CD4 + T cells can provide insight into immunological alterations, predict biomarkers of response to immunotherapy, and guide clinical decision-making in those tumors where the kinetics of the response differ.
Collapse
Affiliation(s)
- Yun Lu
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Hailey A Houson
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Carlos A Gallegos
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | | | - Fang Jia
- ImaginAb, Inc, Inglewood, CA, 90301, USA
| | | | - Patrick N Song
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Shannon E Lynch
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Tiara S Napier
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Ameer Mansur
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Benjamin M Larimer
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Suzanne E Lapi
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Ariella B Hanker
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Anna G Sorace
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
- Departments of Radiology and Biomedical Engineering, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Small Animal Imaging Facility, 1670 University Blvd, Birmingham, USA.
| |
Collapse
|
4
|
Rainone P, Valtorta S, Villa C, Todde S, Cadamuro M, Bertoli G, Conconi D, Lavitrano M, Moresco RM. Evaluating [ 18F]FDG and [ 18F]FLT Radiotracers as Biomarkers of Response for Combined Therapy Outcome in Triple-Negative and Estrogen-Receptor-Positive Breast Cancer Models. Int J Mol Sci 2023; 24:14124. [PMID: 37762429 PMCID: PMC10532149 DOI: 10.3390/ijms241814124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/30/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Breast cancer (BC) is the most frequent cancer and the second leading cause of death in women. A typical feature of BC cells is the metabolic shift toward increased glycolysis, which has become an interesting therapeutic target for metabolic drugs such as metformin (MET). Recently, the administration of the antihypertensive syrosingopine (SYRO) in combination with MET has shown a synergistic effect toward a variety of cancers. However, a fundamental need remains, which is the development of in vivo biomarkers that are able to detect early clinical response. In this study, we exploited a triple-negative murine BC cell line (4T1) and a metastatic ER+ murine BC cell line (TS/A) in order to investigate, in vivo, the early response to treatment, based on MET and/or SYRO administration, evaluating [18F]FDG and [18F]FLT as potential biomarkers via PET/CT. The study provides evidence that SYRO plus MET has a synergistic effect on tumor growth inhibition in both 4T1 and TS/A experimental models and has showed the highest efficacy on the TNBC xenograft mice (4T1) via the expression reduction in the lactate transporter MCT4 and in the epithelial-mesenchymal transition biomarker Snail, promoting its potential application in therapy settings. In addition, the selective reduction in the [18F]FLT tumor uptake (at 7 dd), observed in the SYRO plus MET treated mice in comparison with the vehicle group, suggests that this radiotracer could be potentially used as a biomarker for the early detection of therapy response, in both evaluated xenografts models.
Collapse
Affiliation(s)
- Paolo Rainone
- Department of Medicine and Surgery, University of Milano—Bicocca, 20900 Monza, Italy; (P.R.); (C.V.); (S.T.); (D.C.); (M.L.)
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy;
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 20054 Segrate, Italy;
| | - Silvia Valtorta
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy;
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 20054 Segrate, Italy;
- NBFC National Biodiversity Future Center, 90133 Palermo, Italy
| | - Chiara Villa
- Department of Medicine and Surgery, University of Milano—Bicocca, 20900 Monza, Italy; (P.R.); (C.V.); (S.T.); (D.C.); (M.L.)
| | - Sergio Todde
- Department of Medicine and Surgery, University of Milano—Bicocca, 20900 Monza, Italy; (P.R.); (C.V.); (S.T.); (D.C.); (M.L.)
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 20054 Segrate, Italy;
- Tecnomed Foundation, University of Milano—Bicocca, 20126 Monza, Italy
| | - Massimiliano Cadamuro
- Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy;
- General Internal Medicine Unit, Padua University-Hospital, 35128 Padua, Italy
| | - Gloria Bertoli
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 20054 Segrate, Italy;
- NBFC National Biodiversity Future Center, 90133 Palermo, Italy
| | - Donatella Conconi
- Department of Medicine and Surgery, University of Milano—Bicocca, 20900 Monza, Italy; (P.R.); (C.V.); (S.T.); (D.C.); (M.L.)
| | - Marialuisa Lavitrano
- Department of Medicine and Surgery, University of Milano—Bicocca, 20900 Monza, Italy; (P.R.); (C.V.); (S.T.); (D.C.); (M.L.)
| | - Rosa Maria Moresco
- Department of Medicine and Surgery, University of Milano—Bicocca, 20900 Monza, Italy; (P.R.); (C.V.); (S.T.); (D.C.); (M.L.)
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy;
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 20054 Segrate, Italy;
| |
Collapse
|
5
|
Davenport AA, Lu Y, Gallegos CA, Massicano AVF, Heinzman KA, Song PN, Sorace AG, Cogan NG. Mathematical Model of Triple-Negative Breast Cancer in Response to Combination Chemotherapies. Bull Math Biol 2022; 85:7. [PMID: 36542180 DOI: 10.1007/s11538-022-01108-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 11/14/2022] [Indexed: 12/24/2022]
Abstract
Triple-negative breast cancer (TNBC) is a heterogenous disease that is defined by its lack of targetable receptors, thus limiting treatment options and resulting in higher rates of metastasis and recurrence. Combination chemotherapy treatments, which inhibit tumor cell proliferation and regeneration, are a major component of standard-of-care treatment of TNBC. In this manuscript, we build a coupled ordinary differential equation model of TNBC with compartments that represent tumor proliferation, necrosis, apoptosis, and immune response to computationally describe the biological tumor affect to a combination of chemotherapies, doxorubicin (DRB) and paclitaxel (PTX). This model is parameterized using longitudinal [18F]-fluorothymidine positron emission tomography (FLT-PET) imaging data which allows for a noninvasive molecular imaging approach to quantify the tumor proliferation and tumor volume measurements for two murine models of TNBC. Animal models include a human cell line xenograft model, MDA-MB-231, and a syngeneic 4T1 mammary carcinoma model. The mathematical models are parameterized and the percent necrosis at the end time point is predicted and validated using histological hematoxylin and eosin (H&E) data. Global Sobol' sensitivity analysis is conducted to further understand the role each parameter plays in the model's goodness of fit to the data. In both the MDA-MB-231 and the 4T1 tumor models, the designed mathematical model can accurately describe both tumor volume changes and final necrosis volume. This can give insight into the ordering, dosing, and timing of DRB and PTX treatment. More importantly, this model can also give insight into future novel combinations of therapies and how the immune system plays a role in therapeutic response to TNBC, due to its calibration to two types of TNBC murine models.
Collapse
Affiliation(s)
- Angelica A Davenport
- Department of Mathematics, Florida State University, 1017 Academic Way, Tallahassee, FL, 32304, USA.
| | - Yun Lu
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Carlos A Gallegos
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Katherine A Heinzman
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Patrick N Song
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anna G Sorace
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - N G Cogan
- Department of Mathematics, Florida State University, 1017 Academic Way, Tallahassee, FL, 32304, USA
| |
Collapse
|
6
|
A novel ribociclib derivative WXJ-103 exerts anti-breast cancer effect through CDK4/6. Anticancer Drugs 2022:00001813-990000000-00144. [PMID: 36729405 PMCID: PMC10344442 DOI: 10.1097/cad.0000000000001475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The triple-negative breast cancer (TNBC) subtype is the most aggressive type of breast cancer with a low survival prognosis and high recurrence rate. There is currently no effective treatment to improve it. In this work, we explored the effect of a synthetic compound named WXJ-103 on several aspects of TNBC biology. The human breast cancer cell lines MDA-MB-231 and MCF-7 were used in the experiments, and the cell viability was detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide method, and the cell migration and invasion abilities were detected by wound healing assay and Transwell invasion assay. Cell cycle and apoptosis experiments were analyzed by flow cytometry, and protein levels related to cyclin-dependent kinase (CDK) 4/6-cyclin D-Rb-E2F pathway were analyzed by western blotting. Then, in-vivo experiments were performed to determine the clinical significance and functional role of WXJ-103. The results show that WXJ-103 can inhibit the adhesion, proliferation, migration, and invasion of TNBC cells, and can arrest the cell cycle in G1 phase. The levels of CDK4/6-cyclin D-Rb-E2F pathway-related proteins such as CDK6 and pRb decreased in a dose-dependent manner. Therefore, the antitumor activity of WXJ-103 may depend on the inhibition of CDK4/6-cyclin D1-Rb-E2F pathway. This research shows that WXJ-103 may be a new promising antitumor drug, which can play an antitumor effect on TNBC and provide new ideas for the treatment of TNBC.
Collapse
|
7
|
Lu Y, Massicano AVF, Gallegos CA, Heinzman KA, Parish SW, Warram JM, Sorace AG. Evaluating the Accuracy of FUCCI Cell Cycle In Vivo Fluorescent Imaging to Assess Tumor Proliferation in Preclinical Oncology Models. Mol Imaging Biol 2022; 24:898-908. [PMID: 35650411 DOI: 10.1007/s11307-022-01739-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 12/29/2022]
Abstract
PURPOSE The primary goal of this study is to evaluate the accuracy of the fluorescence ubiquitination cell cycle indicator (FUCCI) system with fluorescence in vivo imaging compared to 3'-deoxy-3'-[18F]fluorothymidine ([18F]-FLT) positron emission tomography (PET)/computed tomography (CT) and biological validation through histology. Imaging with [18F]-FLT PET/CT can be used to noninvasively assess cancer cell proliferation and has been utilized in both preclinical and clinical studies. However, a cost-effective and straightforward method for in vivo, cell cycle targeted cancer drug screening is needed prior to moving towards translational imaging methods such as PET/CT. PROCEDURES In this study, fluorescent MDA-MB-231-FUCCI tumor growth was monitored weekly with caliper measurements and fluorescent imaging. Seven weeks post-injection, [18F]-FLT PET/CT was performed with a preclinical PET/CT, and tumors samples were harvested for histological analysis. RESULTS RFP fluorescent signal significantly correlated with tumor volume (r = 0.8153, p < 0.0001). Cell proliferation measured by GFP fluorescent imaging was correlated with tumor growth rate (r = 0.6497, p < 0.001). Also, GFP+ cells and [18F]-FLT regions of high uptake were both spatially located in the tumor borders, indicating that the FUCCI-IVIS method may provide an accurate assessment of tumor heterogeneity of cell proliferation. The quantification of total GFP signal was correlated with the sum of tumor [18F]-FLT standard uptake value (SUV) (r = 0.5361, p = 0.0724). Finally, histological analysis confirmed viable cells in the tumor and the correlation of GFP + and Ki67 + cells (r = 0.6368, p = 0.0477). CONCLUSION Fluorescent imaging of the cell cycle provides a noninvasive accurate depiction of tumor progression and response to therapy, which may benefit in vivo testing of novel cancer therapeutics that target the cell cycle.
Collapse
Affiliation(s)
- Yun Lu
- Department of Radiology, University of Alabama at Birmingham, Volker Hall G082, 1670 University Boulevard, Birmingham, AL, 35233, USA
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Adriana V F Massicano
- Department of Radiology, University of Alabama at Birmingham, Volker Hall G082, 1670 University Boulevard, Birmingham, AL, 35233, USA
| | - Carlos A Gallegos
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Katherine A Heinzman
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Sean W Parish
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Jason M Warram
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Anna G Sorace
- Department of Radiology, University of Alabama at Birmingham, Volker Hall G082, 1670 University Boulevard, Birmingham, AL, 35233, USA.
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| |
Collapse
|
8
|
Tatum JL, Kalen JD, Jacobs PM, Riffle LA, James A, Thang L, Sanders C, Hollingshead MG, Basuli F, Shi J, Doroshow JH. 3'-[ 18F]fluoro-3'-deoxythymidine ([ 18F]FLT) Positron Emission Tomography as an In Vivo Biomarker of inhibition of CDK 4/6-Rb pathway by Palbociclib in a patient derived bladder tumor. J Transl Med 2022; 20:375. [PMID: 35982453 PMCID: PMC9389794 DOI: 10.1186/s12967-022-03580-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/07/2022] [Indexed: 11/15/2022] Open
Abstract
Background Several new generation CDK4/6 inhibitors have been developed and approved for breast cancer therapy in combination with endocrine therapeutics. Application of these inhibitors either alone or in combination in other solid tumors has been proposed, but no imaging biomarkers of response have been reported in non-breast cancer animal models. The purpose of this study was to evaluate 3'-[18F]fluoro-3'-deoxythymidine ([18F]FLT) Positron Emission Tomography (PET) as in vivo biomarker of response to palbociclib in a non-breast cancer model. Methods Twenty-four NSG mice bearing patient derived xenografts (PDX) of a well-characterized bladder tumor were randomized into 4 treatment groups: vehicle (n = 6); palbociclib (n = 6); temozolomide (n = 6); and palbociclib plus temozolomide (n = 6) and treated with two cycles of therapy or vehicle. Tumor uptake of [18F]FLT was determined by micro-PET/CT at baseline, 3 days, and 9 days post initiation of therapy. Following the second cycle of therapy, the mice were maintained until their tumors reached a size requiring humane termination. Results [18F]FLT uptake decreased significantly in the palbociclib and combination arms (p = 0.0423 and 0.0106 respectively at day 3 and 0.0012 and 0.0031 at day 9) with stable tumor volume. In the temozolomide arm [18F]FLT uptake increased with day 9 uptake significantly different than baseline (p = 0.0418) and progressive tumor growth was observed during the treatment phase. All groups exhibited progressive disease after day 22, 10 days following cessation of therapy. Conclusion Significant decreases in [18F]FLT uptake as early as three days post initiation of therapy with palbociclib, alone or in combination with temozolomide, in this bladder cancer model correlates with an absence of tumor growth during therapy that persists until day 18 for the palbociclib group and day 22 for the combination group (6 days and 10 days) following cessation of therapy. These results support early modulation of [18F]FLT as an in vivo biomarker predictive of palbociclib therapy response in a non-breast cancer model. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03580-8.
Collapse
Affiliation(s)
- James L Tatum
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Joseph D Kalen
- Small Animal Imaging Program, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Paula M Jacobs
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.
| | - Lisa A Riffle
- Small Animal Imaging Program, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Amy James
- Animal Research Technical Support, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Lai Thang
- Animal Research Technical Support, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Chelsea Sanders
- Animal Research Technical Support, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Melinda G Hollingshead
- Biological Testing Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institute of Health, Frederick, MD, United States
| | - Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jianfeng Shi
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
9
|
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
|
10
|
Liu Y, Zhou Q, Song S, Tang S. Integrating metabolic reprogramming and metabolic imaging to predict breast cancer therapeutic responses. Trends Endocrinol Metab 2021; 32:762-775. [PMID: 34340886 DOI: 10.1016/j.tem.2021.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/26/2021] [Accepted: 07/04/2021] [Indexed: 01/10/2023]
Abstract
Metabolic reprogramming is not only an emerging hallmark of cancer, but also an essential regulator of cancer cell adaptation to the microenvironment. Metabolic imaging targeting metabolic signatures has been widely used for breast cancer diagnosis. However, limited implications have been explored for monitoring breast cancer therapy response, although metabolic plasticity is notably associated with therapy resistance. In this review, we focus on the metabolic alterations upon breast cancer therapy and their potential for evaluating breast cancer therapeutic responses. We summarize the metabolic network and regulatory changes upon breast cancer therapy in terms of cancer pathological and genetic differences and discuss the implications of metabolic imaging with various probes in selecting target beneficiaries for precision treatment.
Collapse
Affiliation(s)
- Yi Liu
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China; Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China
| | - Qian Zhou
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China.
| | - Shuang Tang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China; Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China; Shanghai Key Laboratory of Radiation Oncology, Shanghai 201321, PR China.
| |
Collapse
|