1
|
Kenawy MA, Abdelhafez HM, Al-Fatlawi M, Jassim TN, Jasim AS, Alashkar EM. Comparing texture analysis of pretreatment 18F-FDG PET and 68Ga-PSMA PET in patients with prostate cancer: investigation of diagnostic efficacy and prognostic biomarker. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2025; 64:253-261. [PMID: 40137954 DOI: 10.1007/s00411-025-01114-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 02/12/2025] [Indexed: 03/29/2025]
Abstract
This study aims to evaluate the predictive accuracy of textural parameters and current parameters of 18F-fluorodeoxyglucose and 68Ga-labeled prostate-specific antigen positron emission tomography (FDG and PSMA PET) images in prostate cancer (PCa) and compare the features retrieved from both scans. Based on symptoms, digital rectal examination (DRE), prostate-specific antigen (PSA) level in the blood, or histopathology from transrectal ultrasound-guided biopsy and 4Kscore Test, 120 patients have confirmed PCa. Sixty of them were scanned on a PET/CT machine using 18F-FDG, and the other 60 patients were scanned using 68Ga-PSMA of radiopharmacy. Each tumour was delineated using PET. Edge texture parameters were used to define each tumour, and 73 features in all were taken from eight distinct texture matrices and computed using the open-source program Chang-Gung Image Texture Analysis (CGITA). Using Spearman correlation, feature correlation with conventional quantitative metrics (Maximum Standardized Uptake Value (SUVmax), Total Lesion Glycolysis (TLG), Metabolic Tumor Volume (MTV)) was investigated, and it was found that the High-Intensity Low-Energy Radiation (HILRE) correlation was strong. PCa was best discriminated by HILRE (64-bin) in receiver operating characteristic curves. It is concluded that 68Ga-PSMA-based PET imaging is better than 18F-FDG-based PET and is strongly associated with PCa tumour allocation. According to extracted features, HILRE is the most significant measure and it is, thus, considered here an independent predictor of PCa prognosis. Although the study's findings are helpful, confirmation by further prospective research is required.
Collapse
Affiliation(s)
- Mahmoud A Kenawy
- Biophysics Branch, Department of Physics, Faculty of Science (for Boys), Al-Azhar University, Nasr City, Cairo, 11884, Egypt.
- Radiology Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq.
| | - Hussein M Abdelhafez
- Biophysics Branch, Department of Physics, Faculty of Science (for Boys), Al-Azhar University, Nasr City, Cairo, 11884, Egypt
| | - Murtadha Al-Fatlawi
- Radiology Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| | - Tariq Nadhim Jassim
- Radiology Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| | - Ahmed Salman Jasim
- Radiology Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| | - Elsayed M Alashkar
- Biophysics Branch, Department of Physics, Faculty of Science (for Boys), Al-Azhar University, Nasr City, Cairo, 11884, Egypt
| |
Collapse
|
2
|
Chatta R, Shah H, Pallod S. PSMA PET/CT in Detecting Esophageal Cancer and Hepatocellular Carcinoma. A Report of Two Cases. Clin Nucl Med 2025:00003072-990000000-01656. [PMID: 40247467 DOI: 10.1097/rlu.0000000000005901] [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: 01/07/2025] [Accepted: 03/13/2025] [Indexed: 04/19/2025]
Abstract
Prostate-specific membrane antigen (PSMA) PET/CT has improved the diagnosis, staging, and detection of recurrent and metastatic prostate cancer. Nevertheless, PSMA uptake has also been observed in non-prostatic conditions, which underscores the need to explore its broader applications and refine our understanding of its specificity in diverse clinical contexts. In cases of uptake in atypical locations, further workup is warranted to avoid premature conclusions of metastatic prostate disease. We report two cases of PSMA PET uptake: the first involves esophageal cancer, and the second details two synchronous malignancies-hepatocellular and esophageal cancer.
Collapse
Affiliation(s)
- Rami Chatta
- Department of Radiology, Harvard Medical School, Brigham and Women's Hospital
| | - Hina Shah
- Department of Radiology, Harvard Medical School, Brigham and Women's Hospital
| | - Saurabh Pallod
- Department of Nuclear and Molecular Imaging, Harvard Medical School, Dana Farber Cancer Institute, Boston MA
| |
Collapse
|
3
|
Wang L, Ren S, Lou J, Xue S, Zhou P, Zheng X, Shan F, Li X, Chen Y, Liu X. E-Urea-K-Based PSMA Imaging Served as an Alternative in Assessing Tumor Neovascularization via Targeting CD31. Mol Pharm 2025; 22:2029-2039. [PMID: 40013667 DOI: 10.1021/acs.molpharmaceut.4c01252] [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: 02/28/2025]
Abstract
To reveal the natural correlation between prostate-specific membrane antigen (PSMA) imaging and tumor neovascularization in prostate cancer and further explore E-urea-K-based PSMA-targeted (EK-PSMA) imaging as a potential indicator of tumor neovascularization, the 22Rv1 mouse models were established and underwent 99mTc-HYNIC-ALUG SPECT/CT. Pearson correlation analysis was applied to assess the relationship between tumor tracer uptake and tumor characteristics, including size, glucose metabolism, and cell phenotypes (e.g., Ki-67, VEGF, CD31, and PSMA). Then, molecular docking further identified the key factors of EK-PSMA imaging, specifically related to tumor neovascularization. Finally, animal models with positive and negative PSMA expression (22Rv1, LNCaP, U87, SAOS-2, A549, and ACHN) were subjected to antibody-targeted blockade to verify the role of these key factors in EK-PSMA imaging. The Pearson's r values of tracer uptake correlated with CD31 and tumor size were 0.82 and 0.99, respectively (P < 0.05), and the correlations of tracer uptake with SUVmax, SUVmean, Ki-67, VEGF, and PSMA expressions were 0.47, 0.20, 0.69, -0.65, and 0.20, respectively (all P > 0.05). Molecular docking confirmed the affinity of E-urea-K to PSMA (two sites, binding scores, -5.4 kcal/mol and -6.0 kcal/mol) and CD31 (one site, binding score, -5.1 kcal/mol). The blockade of the CD31 antibody partially reduced the 99mTc-HYNIC-ALUG uptake in five other types of tumors (paired t test, P = 0.0478). The Pearson's r value of CD31 staining and tracer uptake prior to the antibody blockade was 0.84 (P < 0.05). Additionally, when removing the PSMA-positive models (22Rv1 and LNCaP), the Pearson's r value of CD31 staining and tracer uptake prior to the antibody blockade was 0.99 (P < 0.05). Thus, CD31 was found to be a mutual target of EK-PSMA imaging; therefore, EK-PSMA imaging provides a viable assessment option for tumor neovascularization, especially for PSMA-negative tumors.
Collapse
Affiliation(s)
- Lan Wang
- Department of Nuclear Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
- Department of Nuclear Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Shengnan Ren
- Department of Nuclear Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Jingjing Lou
- Department of Nuclear Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Shuai Xue
- Department of Nuclear Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Pan Zhou
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Xiaobei Zheng
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Fengling Shan
- Department of Nuclear Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Xiao Li
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
- Department of Radiology, The First Affiliated Hospital of Xiamen University, Xiamen 361000, China
| | - Yangchun Chen
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai 200433, China
| | - Xingdang Liu
- Department of Nuclear Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
- Department of Nuclear Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
4
|
Liu F, Xiao L, Zhao L, Tao Y, Huang D, Chen Z, He C, Wu C. Prostate-specific membrane antigen-targeting radiopharmaceuticals: a new frontier in hepatic malignancies. Front Oncol 2025; 15:1547459. [PMID: 40123907 PMCID: PMC11926431 DOI: 10.3389/fonc.2025.1547459] [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: 12/18/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
Background/Objectives Prostate-specific membrane antigen (PSMA) is overexpressed in prostate hypercellularity, making it an effective target for molecular imaging and therapy of prostate cancer. PSMA is expressed in the neovasculature of hepatic malignancies and regulates tumor cell invasion and angiogenesis. The diagnosis and treatment of hepatic malignancies remain challenging. Thus, radiopharmaceuticals targeting PSMA are gaining prominence in the treatment of hepatic malignancies. Therefore, this review aims to discuss the applications of PSMA-targeting radiopharmaceuticals in hepatic malignant tumors, focusing on hepatocellular carcinoma (HCC), to assess their value as a diagnostic and therapeutic agent for hepatic malignancies. Methods The potentials of PSMA-targeting radiopharmaceuticals for diagnostic and therapeutic use in hepatic malignancies were investigated. Moreover, their characteristics, diagnostic and therapeutic efficacies, and potential synergies when used in conjunction with other therapeutic modalities were elucidated. Results Computed tomography (CT) and magnetic resonance imaging (MRI) are the most common imaging modalities in clinical practice; however, their sensitivity is not optimal. PSMA positron emission tomography/CT can be used as a complementary modality to conventional imaging for characterizing lesions, staging and/or re-staging HCC, and assessing treatment response when conventional imaging results are unclear. Moreover, most patients with HCC are diagnosed at an advanced stage in which treatment options are limited. Hence, PSMA-based radioligand therapy serves as a promising alternative treatment when multiple treatments fail. Conclusions Further research and clinical transformation are required to effectively diagnose and treat HCC via PSMA targeting. This will have significant clinical application prospects in primary and secondary hepatic malignancies.
Collapse
Affiliation(s)
- Fucen Liu
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Mianyang, China
- Department of Nuclear Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Liming Xiao
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Mianyang, China
- Department of Nuclear Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Ling Zhao
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Mianyang, China
- Department of Nuclear Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- Institute of Basic Medicine, North Sichuan Medical College, Nanchong, China
| | - Yi Tao
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Mianyang, China
- Department of Nuclear Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Dan Huang
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Mianyang, China
- Department of Nuclear Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Zhengguo Chen
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Mianyang, China
- Department of Nuclear Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Chuandong He
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Mianyang, China
- Department of Nuclear Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Chunyan Wu
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Mianyang, China
- Department of Nuclear Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| |
Collapse
|
5
|
Khaleel S, Perera M, Papa N, Kuo F, Golkaram M, Rappold P, Kotecha RR, Coleman J, Russo P, Motzer R, Reznik E, Hakimi AA. Gene expression of prostate-specific membrane antigen (FOLH1) in clear cell renal cell carcinoma predicts angiogenesis and response to tyrosine kinase inhibitors. Urol Oncol 2025; 43:192.e21-192.e28. [PMID: 39537440 PMCID: PMC11875958 DOI: 10.1016/j.urolonc.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/28/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE Combination systemic therapies (CSTs) of immuno-oncologic (IO) and VEGF-inhibiting agents (VEGFi) have become the standard of care for management of metastatic clear cell renal cell carcinoma (m-ccRCC). However, treatment outcomes vary between patients, with no established biomarkers to determine optimal CST regimens (IO/IO or IO/VEGFi). Prostate Specific Membrane Antigen (PSMA), encoded by the FOLH1 gene, is a marker of tumor neovasculature in ccRCC, the downstream target of VEGFi. We evaluated the relation between FOLH1 expression and angiogenesis, as well as clinical outcomes, in 5 m-ccRCC ST trials. MATERIALS AND METHODS using Spearman's rank correlation (SPRC) test, we assessed the correlation between FOLH1 expression and gene expression signature (GES) scores corresponding to angiogenic and immunologic features of the tumor microenvironment (TME) of m-ccRCC in our trial cohorts. Using Cox proportional hazard regression (Cox-PHR), we assessed the association between FOLH1 expression level, summarized by within-study quantiles (qFOLH1), and progression-free and overall survival (PFS, OS). RESULTS Increased FOLH1 expression was significantly associated with higher TME angiogenesis GES scores (SPRC +0.5, P < 0.001), but did not consistently correlate with immune feature GES scores. Meta-analysis of PFS in the sunitinib TKI arm of trial cohorts showed an overall positive association with qFOLH1 (HR = 0.89; 95% CI = 0.85-0.94, P < 0.0001). qFOLH1 was not significantly associated with OS in the sunitinib arms of the two trials with OS data (COMPARZ, HR 0.87, 95% CI 0.71-1.07, P = 0.17; and Checkmate-214, HR 0.89, 95% CI 0.67-1.17, P = 0.70). CONCLUSIONS PSMA-encoding FOLH1 gene expression correlates with neoangiogenesis and predicts PFS in m-ccRCC patients treated with sunitinib TKI, suggesting that PSMA PET could be explored as a noninvasive biomarker for guiding CST choice (IO/IO or IO/VEGFi) as well as prediction of treatment response to VEGFi in m-ccRCC patients.
Collapse
Affiliation(s)
- Sari Khaleel
- Minimally Invasive Urology Institute, The Miriam Hospital, Providence, RI; Warren Alpert Medical School of Brown University, Providence, RI
| | - Marlon Perera
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Department of Surgery, Austin Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Nathan Papa
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Fengshen Kuo
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Phillip Rappold
- Department of Urology, University of Rochester Medical Center (URMC), Rochester, NY
| | - Ritesh R Kotecha
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jonathan Coleman
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Paul Russo
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Robert Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ed Reznik
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - A Ari Hakimi
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY.
| |
Collapse
|
6
|
Puik JR, Le C, Kazemier G, Oprea-Lager DE, Swijnenburg RJ, Giovannetti E, Griffioen AW, Huijbers EJ. Prostate-specific membrane antigen as target for vasculature-directed therapeutic strategies in solid tumors. Crit Rev Oncol Hematol 2025; 205:104556. [PMID: 39551117 DOI: 10.1016/j.critrevonc.2024.104556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/21/2024] [Accepted: 11/07/2024] [Indexed: 11/19/2024] Open
Abstract
Prostate-specific membrane antigen (PSMA) is one of the few biomarkers which has been successfully translated to the clinic as theranostic biomarker for patients with prostate cancer. In the context of prostate cancer, PSMA is overexpressed on the cell membrane of tumor cells, making it a viable target for interventions with urea-based small molecule inhibitors or antibodies conjugated to radioactive isotopes. Interestingly, in several non-prostatic cancers, expression of PSMA appears to be associated with the tumor neovasculature. This offers novel therapeutic opportunities for treatments targeting the vasculature in non-prostatic cancers. In this review, we discuss PSMA and its potential as target for vasculature-directed therapeutic approaches, including radioligand therapy, fusion protein vaccination and CAR T-cell therapy.
Collapse
Affiliation(s)
- Jisce R Puik
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Surgery, De Boelelaan 1117, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands
| | - Chung Le
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Geert Kazemier
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Surgery, De Boelelaan 1117, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands
| | - Daniela E Oprea-Lager
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Rutger-Jan Swijnenburg
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Surgery, De Boelelaan 1117, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands
| | - Elisa Giovannetti
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands; Cancer Pharmacology Lab, Associazione Italiana per la Ricerca sul Cancro (AIRC), Fondazione Pisana per la Scienza, University of Pisa, Pisa, Italy.
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; CimCure BV, Plesmanlaan 125, Amsterdam, the Netherlands
| | - Elisabeth Jm Huijbers
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; CimCure BV, Plesmanlaan 125, Amsterdam, the Netherlands
| |
Collapse
|
7
|
Skubal M, Larney BM, Phung NB, Desmaras JC, Dozic AV, Volpe A, Ogirala A, Machado CL, Djibankov J, Ponomarev V, Grimm J. Vascularized tumor on a microfluidic chip to study mechanisms promoting tumor neovascularization and vascular targeted therapies. Theranostics 2025; 15:766-783. [PMID: 39776800 PMCID: PMC11700857 DOI: 10.7150/thno.95334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 11/05/2024] [Indexed: 01/11/2025] Open
Abstract
The cascade of events leading to tumor formation includes induction of a tumor supporting neovasculature, as a primary hallmark of cancer. Developing vasculature is difficult to evaluate in vivo but can be captured using microfluidic chip technology and patient derived cells. Herein, we established an on chip approach to investigate the mechanisms promoting tumor vascularization and vascular targeted therapies via co-culture of cancer spheroids and endothelial cells in a three dimensional environment. Methods: We investigated both, tumor neovascularization and therapy, via co-culture of human derived endothelial cells and adjacently localized metastatic renal cell carcinoma spheroids on a commercially available microfluidic chip system. Metastatic renal cell carcinoma spheroids adjacent to primary vessels model tumor, and induce vessels to sprout neovasculature towards the tumor. We monitored real time changes in vessel formation, probed the interactions of tumor and endothelial cells, and evaluated the role of important effectors in tumor vasculature. In addition to wild type endothelial cells, we evaluated endothelial cells that overexpress Prostate Specific Membrane Antigen (PSMA), that has emerged as a marker of tumor associated neovasculature. We characterized the process of neovascularization on the microfluidic chip stimulated by enhanced culture medium and the investigated metastatic renal cell carcinomas, and assessed endothelial cells responses to vascular targeted therapy with bevacizumab via confocal microscopy imaging. To emphasize the potential clinical relevance of metastatic renal cell carcinomas on chip, we compared therapy with bevacizumab on chip with an in vivo model of the same tumor. Results: Our model permitted real-time, high-resolution observation and assessment of tumor-induced angiogenesis, where endothelial cells sprouted towards the tumor and mimicked a vascular network. Bevacizumab, an antiangiogenic agent, disrupted interactions between vessels and tumors, destroying the vascular network. The on chip approach enabled assessment of endothelial cell biology, vessel's functionality, drug delivery, and molecular expression of PSMA. Conclusion: Observations in the vascularized tumor on chip permitted direct and conclusive quantification of vascular targeted therapies in weeks as opposed to months in a comparable animal model, and bridged the gap between in vitro and in vivo models.
Collapse
Affiliation(s)
- Magdalena Skubal
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Benedict Mc Larney
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ngan Bao Phung
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA
| | - Juan Carlos Desmaras
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Abdul Vehab Dozic
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alessia Volpe
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anuja Ogirala
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Camila Longo Machado
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jakob Djibankov
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vladimir Ponomarev
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | - Jan Grimm
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
8
|
Bogdanović B, Fagret D, Ghezzi C, Montemagno C. Integrin Targeting and Beyond: Enhancing Cancer Treatment with Dual-Targeting RGD (Arginine-Glycine-Aspartate) Strategies. Pharmaceuticals (Basel) 2024; 17:1556. [PMID: 39598465 PMCID: PMC11597078 DOI: 10.3390/ph17111556] [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: 09/30/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
Integrins, an important superfamily of cell adhesion receptors, play an essential role in cancer progression, metastasis, and angiogenesis, establishing them as prime targets for both diagnostic and therapeutic applications. Despite their significant potential, integrin-targeted therapies have faced substantial challenges in clinical trials, including variable efficacy and unmet high expectations. Nevertheless, the consistent expression of integrins on tumor and stromal cells underscores their ongoing relevance and potential. Traditional RGD-based imaging and therapeutic agents have faced limitations, such as inconsistent target expression and rapid systemic clearance, which have reduced their effectiveness. To overcome these challenges, recent research has focused on advancing RGD-based strategies and exploring innovative solutions. This review offers a thorough analysis of the latest developments in the RGD-integrin field, with a particular focus on addressing previous limitations. It delves into new dual-targeting approaches and cutting-edge RGD-based agents designed to improve both tumor diagnosis and therapeutic outcomes. By examining these advancements, this review illuminates new pathways for enhancing the specificity and efficacy of integrin-targeted therapies, paving the way for more effective cancer diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Bojana Bogdanović
- INSERM, CHU Grenoble Alpes, Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, 38000 Grenoble, France; (B.B.); (D.F.); (C.G.)
| | - Daniel Fagret
- INSERM, CHU Grenoble Alpes, Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, 38000 Grenoble, France; (B.B.); (D.F.); (C.G.)
| | - Catherine Ghezzi
- INSERM, CHU Grenoble Alpes, Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, 38000 Grenoble, France; (B.B.); (D.F.); (C.G.)
| | | |
Collapse
|
9
|
Filippi L, Ferrari C, Rubini G. Theranostic strategies in sarcoma: preliminary clinical evidence. Expert Opin Investig Drugs 2024; 33:1119-1127. [PMID: 39367699 DOI: 10.1080/13543784.2024.2414119] [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: 06/10/2024] [Revised: 09/07/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
INTRODUCTION Sarcomas encompass a highly diverse range of malignancies, characterized by varied morphological and molecular profiles. Treatment options in case of therapy-refractory or advanced disease are limited. In this context, theranostics emerges as an innovative platform seamlessly integrating diagnosis and therapy, offering promising prospects. AREAS COVERED This special report delves into the initial clinical applications of theranostic-based approaches in sarcomas. Specifically, it examines various strategies targeting biomarkers associated with sarcomas, including fibroblast activation protein (FAP), prostate-specific membrane antigen (PSMA), C-X-C chemokine receptor type 4 (CXCR4) and somatostatin receptor 2 (SSTR2). EXPERT OPINION The heterogeneous uptake of the CXCR4-targeted radioligand in lesions, along with its poor correlation with immunohistochemistry data, diminishes the attractiveness of this theranostic approach in the sarcoma oncological setting. SSTR2-targeted approaches in sarcoma, although potentially effective, are limited to a single case. Early experiences with FAP inhibitors in sarcoma patients have shown particularly promising outcomes, indicating effective disease control with minimal toxicity. While PSMA presents an enticing target for theranostic approaches in sarcomas, its utilization remains anecdotal and requires further investigation. Prospective and well-designed clinical trials are imperative to delineate the potential impact of FAPI- and PSMA-based approaches on sarcoma therapeutic landscapes, offering innovative and personalized treatment options.
Collapse
Affiliation(s)
- Luca Filippi
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | - Cristina Ferrari
- Nuclear Medicine Unit, Interdisciplinary Department of Medicine (DIM), University of Bari "Aldo Moro", Bari, Italy
| | - Giuseppe Rubini
- Nuclear Medicine Unit, Interdisciplinary Department of Medicine (DIM), University of Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
10
|
Çerçi Koçar İ, Özcan PP, Koç ZP, Süle M, Akbay E, Gen R, Sezer K. Retrospective analysis of thyroid incidentalomas detected by [ 68Ga]Ga-PSMA-11 PET/CT. Endocrine 2024; 86:302-309. [PMID: 38727868 DOI: 10.1007/s12020-024-03847-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/20/2024] [Indexed: 10/02/2024]
Abstract
BACKGROUND Prostate cancer patients, undergo imaging procedures, with [68Ga]Ga-PSMA-11 PET/CT (prostate-specific membrane antigen based positron emission tomography/computed tomography) utilized for primary and secondary staging. PSMA thyroid incidentalomas (PTI) are discovered in the thyroid gland while imaging prostate cancer patients with [68Ga]Ga-PSMA-11 PET/CT. AIMS The aim of the study was to determine the clinical significance of PTIs detected on [68Ga]Ga-PSMA-11 PET/CT. Another goal was to identify a possible threshold for the maximum standardized uptake value (SUVmax), above which a malignant growth could be suspected. STUDY DESIGN A retrospective cross-sectional study. METHODS 769 patients with prostat cancer who underwent [68Ga]Ga-PSMA-11 PET/CT scans in the nuclear medicine department of a tertiary care hospital between January 2020 and December 2022 were retrospectively screened in this study. We analyzed 67 patients in whom PTI was detected. Patients who exceeded the inclusion criteria had their thyroid ultrasonography and ultrasonography -guided fine needle aspiration findings analyzed. RESULTS PTI was discovered in 67 patients (8%). 42 patients who met the inclusion and exclusion criteria were included in the study. Of the 4 malignant patients (9.5%) in the study population, 2 were classified as TIRADS 3 and 2 were classified as TIRADS 4. The cut-off SUVmax value was found to be 5.6. With 100% sensitivity and 47.37% specificity, a cutoff SUVmax of 5.3 was determined through receiver-operator characteristic analysis in order to predict malignant cytology. CONCLUSION PTI is a significant clinical finding; most of diffuse and focal uptakes are frequently related to benign diseases. Each center should establish its own a possible SUVmax cut-off over which a malignant lesion should be suspected.
Collapse
Affiliation(s)
- İlkcan Çerçi Koçar
- Mersin University Medical School, Endocrinology and Metabolism Department, Mersin, Turkey.
- Gaziantep City Educational and Research Hospital, Endocrinology and Metabolism Department, Gaziantep, Turkey.
| | - Pınar Pelin Özcan
- Mersin University Medical School, Nuclear Medicine Department, Mersin, Turkey
| | - Zehra Pınar Koç
- Mersin University Medical School, Nuclear Medicine Department, Mersin, Turkey
| | - Mehmet Süle
- Mersin University Medical School, Endocrinology and Metabolism Department, Mersin, Turkey
| | - Esen Akbay
- Mersin University Medical School, Endocrinology and Metabolism Department, Mersin, Turkey
| | - Ramazan Gen
- Mersin University Medical School, Endocrinology and Metabolism Department, Mersin, Turkey
| | - Kerem Sezer
- Mersin University Medical School, Endocrinology and Metabolism Department, Mersin, Turkey
| |
Collapse
|
11
|
Li RX, Hao Y, Ettel M. Expression of PSMA in Tumor-Associated Vasculature Predicts Poorer Survival in Patients With Hepatocellular Carcinoma and Is Likely Associated With PD-L1. Int J Surg Pathol 2024; 32:1248-1255. [PMID: 38321782 DOI: 10.1177/10668969241226705] [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: 02/08/2024]
Abstract
BACKGROUND PSMA (prostate-specific membrane antigen) is a type II transmembrane glycoprotein recently found to be expressed in hepatocellular carcinoma (HCC). We aimed to characterize the expression pattern of PSMA in HCC and its association with clinicopathologic parameters and other biomarkers. METHODS Immunohistochemical studies for PSMA were performed on a previously established tissue microarray of 103 surgically resected HCC. RESULTS Conceivable PSMA expression in ≥5% tumor-associated vasculature (TAV) was considered positive, and was identified in 56 (54.4%) tumors. Eight (7.8%) tumors also showed membranous/cytoplasmic and/or canalicular staining in tumor cells. By chi-square tests, only PSMA-positive TAV was associated with moderate-to-poorly differentiated HCC and the modified higher tumor stage (P < .05). PSMA-positive TAV was not associated with age, sex, or expression of glypican-3, keratin 7, CD3, CD8, HHLA-2, but marginally correlated with programmed death-ligand 1 (PD-L1) expression (P = .052). Kaplan-Meier survival analysis revealed PSMA-positive TAV as an independent risk factor for poorer disease-specific survival (P = .008). Co-expression of PD-L1 did not ameliorate the adverse prognostication of PSMA-positive TAV. Membranous/cytoplasmic/canalicular expression of PSMA alone was not prognostically significant. CONCLUSIONS Our study confirmed that PSMA-positive TAV is a prospective diagnostic and prognostic biomarker for HCC. Co-expression of PSMA with PD-L1 may suggest potential crosstalk between the 2 proteins, likely regulating the tumor microenvironment.
Collapse
Affiliation(s)
- Rena X Li
- High School Intern, Pittsford Mendon High School, Pittsford, NY, USA
| | - Yansheng Hao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Mark Ettel
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
12
|
Shirke AA, Wang J, Ramamurthy G, Mahanty A, Walker E, Zhang L, Panigrahi A, Wang X, Basilion JP. Prostate Specific Membrane Antigen Expression in a Syngeneic Breast Cancer Mouse Model. Mol Imaging Biol 2024; 26:714-728. [PMID: 38760621 PMCID: PMC11281974 DOI: 10.1007/s11307-024-01920-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/09/2024] [Accepted: 04/22/2024] [Indexed: 05/19/2024]
Abstract
PURPOSE Prostate specific membrane antigen (PSMA) has been studied in human breast cancer (BCa) biopsies, however, lack of data on PSMA expression in mouse models impedes development of PSMA-targeted therapies, particularly in improving breast conserving surgery (BCS) margins. This study aimed to validate and characterize the expression of PSMA in murine BCa models, demonstrating that PSMA can be utilized to improve therapies and imaging techniques. METHODS Murine triple negative breast cancer 4T1 cells, and human cell lines, MDA-MB-231, MDA-MB-468, implanted into the mammary fat pads of BALB/c mice, were imaged by our PSMA targeted theranostic agent, PSMA-1-Pc413, and tumor to background ratios (TBR) were calculated to validate selective uptake. Immunohistochemistry was used to correlate PSMA expression in relation to CD31, an endothelial cell biomarker highlighting neovasculature. PSMA expression was also quantified by Reverse Transcriptase Polymerase Chain Reaction (RT-PCR). RESULTS Accumulation of PSMA-1-Pc413 was observed in 4T1 primary tumors and associated metastases. Average TBR of 4T1 tumors were calculated to be greater than 1.5-ratio at which tumor tissues can be distinguished from normal structures-at peak accumulation with the signal intensity in 4T1 tumors comparable to that in high PSMA expressing PC3-pip tumors. Extraction of 4T1 tumors and lung metastases followed by RT-PCR analysis and PSMA-CD31 co-staining shows that PSMA is consistently localized on tumor neovasculature with no expression in tumor cells and surrounding normal tissues. CONCLUSION The selective uptake of PSMA-1-Pc413 in these cancer tissues as well as the characterization and validation of PSMA expression on neovasculature in this syngeneic 4T1 model emphasizes their potential for advancements in targeted therapies and imaging techniques for BCa. PSMA holds great promise as an oncogenic target for BCa and its associated metastases.
Collapse
Affiliation(s)
- Aditi A Shirke
- Department of Biomedical Engineering, Case Western Reserve University, 11100 Euclid Ave, Wearn Building B-49, Cleveland, OH, 44106, USA.
| | - Jing Wang
- Department of Radiology, Case Western Reserve University, 11100 Euclid Ave, Wearn Building B-49, Cleveland, OH, 44106, USA
| | - Gopolakrishnan Ramamurthy
- Department of Radiology, Case Western Reserve University, 11100 Euclid Ave, Wearn Building B-49, Cleveland, OH, 44106, USA
| | - Arpan Mahanty
- Department of Radiology, Case Western Reserve University, 11100 Euclid Ave, Wearn Building B-49, Cleveland, OH, 44106, USA
| | - Ethan Walker
- Department of Biomedical Engineering, Case Western Reserve University, 11100 Euclid Ave, Wearn Building B-49, Cleveland, OH, 44106, USA
| | - Lifang Zhang
- Department of Radiology, Case Western Reserve University, 11100 Euclid Ave, Wearn Building B-49, Cleveland, OH, 44106, USA
| | - Abhiram Panigrahi
- Department of Radiology, Case Western Reserve University, 11100 Euclid Ave, Wearn Building B-49, Cleveland, OH, 44106, USA
| | - Xinning Wang
- Department of Biomedical Engineering, Case Western Reserve University, 11100 Euclid Ave, Wearn Building B-49, Cleveland, OH, 44106, USA.
| | - James P Basilion
- Department of Biomedical Engineering, Case Western Reserve University, 11100 Euclid Ave, Wearn Building B-49, Cleveland, OH, 44106, USA.
- Department of Radiology, Case Western Reserve University, 11100 Euclid Ave, Wearn Building B-49, Cleveland, OH, 44106, USA.
| |
Collapse
|
13
|
Taylor CA, Glover M, Maher J. CAR-T cell technologies that interact with the tumour microenvironment in solid tumours. Expert Rev Clin Immunol 2024; 20:849-871. [PMID: 39021098 DOI: 10.1080/1744666x.2024.2380894] [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: 04/30/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
INTRODUCTION Chimeric antigen receptor (CAR) T-cells have emerged as a ground-breaking therapy for the treatment of hematological malignancies due to their capacity for rapid tumor-specific killing and long-lasting tumor immunity. However, the same success has not been observed in patients with solid tumors. Largely, this is due to the additional challenges imposed by safe and uniform target selection, inefficient CAR T-cell access to sites of disease and the presence of a hostile immunosuppressive tumor microenvironment. AREAS COVERED Literature was reviewed on the PubMed database from the first description of a CAR by Kuwana, Kurosawa and colleagues in December 1987 through to the present day. This literature indicates that in order to tackle solid tumors, CAR T-cells can be further engineered with additional armoring strategies that facilitate trafficking to and infiltration of malignant lesions together with reversal of suppressive immune checkpoints that operate within solid tumor lesions. EXPERT OPINION In this review, we describe a number of recent advances in CAR T-cell technology that set out to combat the problems imposed by solid tumors including tumor recruitment, infiltration, immunosuppression, metabolic compromise, and hypoxia.
Collapse
Affiliation(s)
| | | | - John Maher
- Leucid Bio Ltd, Guy's Hospital, London, UK
- King's College London, School of Cancer and Pharmaceutical Sciences, Guy's Hospital, London, UK
- Department of Immunology, Eastbourne Hospital, Eastbourne, East Sussex, UK
| |
Collapse
|
14
|
Tayara O, Poletajew S, Malewski W, Kunikowska J, Pełka K, Kryst P, Nyk Ł. Prostate-Specific Membrane Antigen Expression in Patients with Primary Prostate Cancer: Diagnostic and Prognostic Value in Positron Emission Tomography-Prostate-Specific Membrane Antigen. Curr Oncol 2024; 31:4165-4177. [PMID: 39195294 PMCID: PMC11352643 DOI: 10.3390/curroncol31080311] [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: 06/24/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/29/2024] Open
Abstract
Prostate cancer represents a significant public health challenge, with its management requiring precise diagnostic and prognostic tools. Prostate-specific membrane antigen (PSMA), a cell surface enzyme overexpressed in prostate cancer cells, has emerged as a pivotal biomarker. PSMA's ability to increase the sensitivity of PET imaging has revolutionized its application in the clinical management of prostate cancer. The advancements in PET-PSMA imaging technologies and methodologies, including the development of PSMA-targeted radiotracers and optimized imaging protocols, led to diagnostic accuracy and clinical utility across different stages of prostate cancer. This highlights its superiority in staging and its comparative effectiveness against conventional imaging modalities. This paper analyzes the impact of PET-PSMA on prostate cancer management, discussing the existing challenges and suggesting future research directions. The integration of recent studies and reviews underscores the evolving understanding of PET-PSMA imaging, marking its significant but still expanding role in clinical practice. This comprehensive review serves as a crucial resource for clinicians and researchers involved in the multifaceted domains of prostate cancer diagnosis, treatment, and management.
Collapse
Affiliation(s)
- Omar Tayara
- Second Department of Urology, Centre of Postgraduate Medical Education, 02-511 Warsaw, Poland; (S.P.); (W.M.); (P.K.); (Ł.N.)
| | - Sławomir Poletajew
- Second Department of Urology, Centre of Postgraduate Medical Education, 02-511 Warsaw, Poland; (S.P.); (W.M.); (P.K.); (Ł.N.)
| | - Wojciech Malewski
- Second Department of Urology, Centre of Postgraduate Medical Education, 02-511 Warsaw, Poland; (S.P.); (W.M.); (P.K.); (Ł.N.)
| | - Jolanta Kunikowska
- Department of Nuclear Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland; (J.K.); (K.P.)
| | - Kacper Pełka
- Department of Nuclear Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland; (J.K.); (K.P.)
- Department of Methodology Laboratory, Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Piotr Kryst
- Second Department of Urology, Centre of Postgraduate Medical Education, 02-511 Warsaw, Poland; (S.P.); (W.M.); (P.K.); (Ł.N.)
| | - Łukasz Nyk
- Second Department of Urology, Centre of Postgraduate Medical Education, 02-511 Warsaw, Poland; (S.P.); (W.M.); (P.K.); (Ł.N.)
| |
Collapse
|
15
|
Bidkar AP, Zerefa L, Yadav S, VanBrocklin HF, Flavell RR. Actinium-225 targeted alpha particle therapy for prostate cancer. Theranostics 2024; 14:2969-2992. [PMID: 38773983 PMCID: PMC11103494 DOI: 10.7150/thno.96403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/01/2024] [Indexed: 05/24/2024] Open
Abstract
Targeted alpha particle therapy (TAT) has emerged as a promising strategy for the treatment of prostate cancer (PCa). Actinium-225 (225Ac), a potent alpha-emitting radionuclide, may be incorporated into targeting vectors, causing robust and in some cases sustained antitumor responses. The development of radiolabeling techniques involving EDTA, DOTA, DOTPA, and Macropa chelators has laid the groundwork for advancements in this field. At the forefront of clinical trials with 225Ac in PCa are PSMA-targeted TAT agents, notably [225Ac]Ac-PSMA-617, [225Ac]Ac-PSMA-I&T and [225Ac]Ac-J591. Ongoing investigations spotlight [225Ac]Ac-hu11B6, [225Ac]Ac-YS5, and [225Ac]Ac-SibuDAB, targeting hK2, CD46, and PSMA, respectively. Despite these efforts, hurdles in 225Ac production, daughter redistribution, and a lack of suitable imaging techniques hinder the development of TAT. To address these challenges and additional advantages, researchers are exploring alpha-emitting isotopes including 227Th, 223Ra, 211At, 213Bi, 212Pb or 149Tb, providing viable alternatives for TAT.
Collapse
Affiliation(s)
- Anil P. Bidkar
- Department of Radiology and Biomedical Imaging, University of California San Francisco, CA-94107, USA
| | - Luann Zerefa
- Department of Radiology and Biomedical Imaging, University of California San Francisco, CA-94107, USA
| | - Surekha Yadav
- Department of Radiology and Biomedical Imaging, University of California San Francisco, CA-94107, USA
| | - Henry F. VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California San Francisco, CA-94107, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA-94107, USA
| | - Robert R. Flavell
- Department of Radiology and Biomedical Imaging, University of California San Francisco, CA-94107, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA-94107, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA-94107, USA
| |
Collapse
|
16
|
Feng YY, Shi YR, Xia Z, Xu L, Li WB, Pang H, Wang ZJ. The clinical signification and application value of [ 68Ga]Ga-PSMA imaging in thyroid malignancy. Endocrine 2024; 84:598-606. [PMID: 37987969 DOI: 10.1007/s12020-023-03599-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/03/2023] [Indexed: 11/22/2023]
Abstract
PURPOSE Approximately 5% of differentiated thyroid cancers lose the ability to uptake iodine, leading to limited treatment options and poor prognosis due to invasion and distant metastasis. PSMA imaging probes have been proposed as a potential diagnostic and therapeutic tool for iodine-refractory thyroid cancer. However, there are limited reports and significant heterogeneity in patient selection, warranting further exploration of the application value of PSMA in thyroid cancer. METHODS We performed Western Blot, PCR, and [68Ga]Ga-PSMA uptake experiments on cell lines and conducted in vivo small animal imaging. Clinical and radiological results of included differentiated thyroid cancer patients were collected. (Trial registration number: 2021-669, Trial registration date: December 30, 2021). RESULTS PSMA expression levels were significantly higher in poorly differentiated thyroid cancer (7.86 ± 1.90 vs. 1.00 ± 0, P < 0.01; 7.86 ± 1.90 vs. 0.03 ± 0.02, P < 0.01), but [68Ga]Ga-PSMA imaging correlated with tumor burden, such as 18F-FDG (8.08 ± 7.74 and 5.67 ± 4.23, P = 0.01) and Tg levels (307.1 ± 183.4 vs. 118.0 ± 116.1, P = 0.002). CONCLUSION Our results showed that PSMA expression increased with the decrease of thyroid cancer differentiation. However, the level of [68Ga]Ga-PSMA uptake in thyroid cancer patients was not significantly associated with the degree of thyroid cancer differentiation, but also with the metabolism and burden of tumors such as 2-[18F]FDG and Tg levels. These findings provide additional clinical significance and application value for PSMA in thyroid cancer.
Collapse
Affiliation(s)
- Yu Yue Feng
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Chongqing, PR China
| | - Yang Rui Shi
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Chongqing, PR China
| | - Zhu Xia
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Chongqing, PR China
| | - Lu Xu
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Chongqing, PR China
| | - Wen Bo Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Chongqing, PR China
| | - Hua Pang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Chongqing, PR China.
| | - Zheng Jie Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Chongqing, PR China.
| |
Collapse
|
17
|
Denmeade SR. Resolute Progress Down a Long and Winding Road Leads to the Promised Land of Prostate-Specific Membrane Antigen-Based Therapies for Prostate Cancer. J Clin Oncol 2024; 42:852-856. [PMID: 38181307 DOI: 10.1200/jco.23.02310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 10/31/2023] [Indexed: 01/07/2024] Open
|
18
|
Bui VN, Unterrainer LM, Brendel M, Kunte SC, Holzgreve A, Allmendinger F, Bartenstein P, Klauschen F, Unterrainer M, Staehler M, Ledderose S. PSMA-Expression Is Highly Associated with Histological Subtypes of Renal Cell Carcinoma: Potential Implications for Theranostic Approaches. Biomedicines 2023; 11:3095. [PMID: 38002095 PMCID: PMC10668989 DOI: 10.3390/biomedicines11113095] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023] Open
Abstract
In renal cell carcinoma (RCC), accurate imaging methods are required for treatment planning and response assessment to therapy. In addition, there is an urgent need for new therapeutic options, especially in metastatic RCC. One way to combine diagnostics and therapy in a so-called theranostic approach is the use of radioligands directed against surface antigens. For instance, radioligands against prostate-specific membrane antigen (PSMA) have already been successfully used for diagnosis and radionuclide therapy of metastatic prostate cancer. Recent studies have demonstrated that PSMA is expressed not only in prostate cancer but also in the neovasculature of several solid tumors, which has raised hopes to use PSMA-guided theranostic approaches in other tumor entities, too. However, data on PSMA expression in different histopathological subtypes of RCC are sparse. Because a better understanding of PSMA expression in RCC is critical to assess which patients would benefit most from theranostic approaches using PSMA-targeted ligands, we investigated the expression pattern of PSMA in different subtypes of RCC on protein level. Immunohistochemical staining for PSMA was performed on formalin-fixed, paraffin-embedded archival material of major different histological subtypes of RCC (clear cell RCC (ccRCC)), papillary RCC (pRCC) and chromophobe RCC (cpRCC). The extent and intensity of PSMA staining were scored semi-quantitatively and correlated with the histological RCC subtypes. Group comparisons were calculated with the Kruskal-Wallis test. In all cases, immunoreactivity was detected only in the tumor-associated vessels and not in tumor cells. Staining intensity was the strongest in ccRCC, followed by cpRCC and pRCC. ccRCC showed the most diffuse staining pattern, followed by cpRCC and pRCC. Our results provide a rationale for PSMA-targeted theranostic approaches in ccRCC and cpRCC.
Collapse
Affiliation(s)
- Vinh Ngoc Bui
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
| | - Lena M. Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Sophie C. Kunte
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
| | - Adrien Holzgreve
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
| | - Fabian Allmendinger
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
| | - Peter Bartenstein
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
| | | | - Marcus Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
- Die RADIOLOGIE, 80331 Munich, Germany
| | - Michael Staehler
- Department of Urology, LMU University Hospital, LMU Munich, 81377 Munich, Germany;
| | - Stephan Ledderose
- Institute of Pathology, LMU Munich, 81377 Munich, Germany; (F.K.); (S.L.)
| |
Collapse
|
19
|
Ritawidya R, Wongso H, Effendi N, Pujiyanto A, Lestari W, Setiawan H, Humani TS. Lutetium-177-Labeled Prostate-Specific Membrane Antigen-617 for Molecular Imaging and Targeted Radioligand Therapy of Prostate Cancer. Adv Pharm Bull 2023; 13:701-711. [PMID: 38022814 PMCID: PMC10676551 DOI: 10.34172/apb.2023.079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 02/04/2023] [Accepted: 04/24/2023] [Indexed: 12/01/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA) represents a promising target for PSMA-overexpressing diseases, especially prostate cancer-a common type of cancer among men worldwide. In response to the challenges in tackling prostate cancers, several promising PSMA inhibitors from a variety of molecular scaffolds (e.g., phosphorous-, thiol-, and urea-based molecules) have been developed. In addition, PSMA inhibitors bearing macrocyclic chelators have attracted interest due to their favorable pharmacokinetic properties. Recently, conjugating a small PSMA molecule inhibitor-bearing 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) chelator, as exemplified by [177Lu]Lu-PSMA-617 could serve as a molecular imaging probe and targeted radioligand therapy (TRT) of metastatic castration resistant prostate cancer (mCRPC). Hence, studies related to mCRPC have drawn global attention. In this review, the recent development of PSMA ligand-617-labeled with 177Lu for the management of mCRPC is presented. Its molecular mechanism of action, safety, efficacy, and future direction are also described.
Collapse
Affiliation(s)
- Rien Ritawidya
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, National Research and Innovation Agency (BRIN), Kawasan Puspiptek, Setu, Tangerang Selatan, 15314 Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Raya Bandung-Sumedang KM 21, Sumedang, 45363, Indonesia
| | - Hendris Wongso
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, National Research and Innovation Agency (BRIN), Kawasan Puspiptek, Setu, Tangerang Selatan, 15314 Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Raya Bandung-Sumedang KM 21, Sumedang, 45363, Indonesia
| | - Nurmaya Effendi
- Faculty of Pharmacy, University of Muslim Indonesia, Kampus II UMI, Jl. Urip Sumoharjo No.225, Panaikang, Panakkukang, Kota, Makassar, Sulawesi Selatan 90231
| | - Anung Pujiyanto
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, National Research and Innovation Agency (BRIN), Kawasan Puspiptek, Setu, Tangerang Selatan, 15314 Indonesia
| | - Wening Lestari
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, National Research and Innovation Agency (BRIN), Kawasan Puspiptek, Setu, Tangerang Selatan, 15314 Indonesia
| | - Herlan Setiawan
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, National Research and Innovation Agency (BRIN), Kawasan Puspiptek, Setu, Tangerang Selatan, 15314 Indonesia
| | - Titis Sekar Humani
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, National Research and Innovation Agency (BRIN), Kawasan Puspiptek, Setu, Tangerang Selatan, 15314 Indonesia
| |
Collapse
|
20
|
Wang JH, Kiess AP. PSMA-targeted therapy for non-prostate cancers. Front Oncol 2023; 13:1220586. [PMID: 37645427 PMCID: PMC10461313 DOI: 10.3389/fonc.2023.1220586] [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: 05/10/2023] [Accepted: 07/25/2023] [Indexed: 08/31/2023] Open
Abstract
Radioligand therapy (RLT) agents are demonstrating a crucial role in the clinical approach to aggressive malignancies such as metastatic castrate-resistant prostate cancer (m-CRPC). With the recent FDA approval of prostate-specific membrane antigen (PSMA)-targeted RLT for m-CRPC, the field has broadened its gaze to explore other cancers that express PSMA in the tumor parenchyma or tumor neovasculature. In this review article, we discuss current progress in the clinical use of PSMA RLTs in non-prostate cancers such salivary gland cancers, renal cell carcinoma, high grade glioma, and soft tissue sarcoma. We highlight early reports in small case series and clinical trials indicating promise for PSMA-targeted RLT and highlighting the importance of identifying patient cohorts who may most benefit from these interventions. Further study is indicated in non-prostate cancers investigating PSMA RLT dosimetry, PSMA PET/CT imaging as a biomarker, and assessing PSMA RLT safety and efficacy in these cancers.
Collapse
Affiliation(s)
- Jarey H. Wang
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Ana P. Kiess
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
21
|
Skubal M, Larney BM, Phung NB, Desmaras JC, Dozic AV, Volpe A, Ogirala A, Machado CL, Djibankov J, Ponomarev V, Grimm J. Vascularized tumor on a microfluidic chip to study mechanisms promoting tumor neovascularization and vascular targeted therapies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.07.552309. [PMID: 37609216 PMCID: PMC10441301 DOI: 10.1101/2023.08.07.552309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
The cascade of events leading to tumor formation includes induction of a tumor supporting neovasculature as a primary hallmark of cancer. Developing vasculature is difficult to evaluate in vivo but can be captured using microfluidic chip technology and patient derived cells. Herein, we established an on chip approach to investigate the mechanisms promoting tumor vascularization and vascular targeted therapies via co-culture of metastatic renal cell carcinoma spheroids and endothelial cells in a 3D environment. Our model permitted real-time, high-resolution observation and assessment of tumor-induced angiogenesis, where endothelial cells sprout towards the tumor and mimic a vascular network. Bevacizumab, an angiogenic inhibitor, disrupted interactions between vessels and tumors, destroying the vascular network. The on chip approach enabled assessment of endothelial cell biology, vessel's functionality, drug delivery, and molecular expression of PSMA. Finally, observations in the vascularized tumor on chip permitted direct and conclusive quantification of this therapy in weeks as opposed to months in a comparable animal model. Teaser Vascularized tumor on microfluidic chip provides opportunity to study targeted therapies and improves preclinical drug discovery.
Collapse
|
22
|
Kuzmanov A, Salemi S, Schmid FA, Burger IA, Eberli D, Kranzbühler B. Improved Prostate-Specific Membrane Antigen (PSMA) Stimulation Using a Super Additive Effect of Dutasteride and Lovastatin In Vitro. Int J Mol Sci 2023; 24:12338. [PMID: 37569712 PMCID: PMC10419009 DOI: 10.3390/ijms241512338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA)-based imaging improved the detection of primary, recurrent and metastatic prostate cancer. However, in certain patients, a low PSMA surface expression can be a limitation for this promising diagnostic tool. Pharmacological induction of PSMA might be useful to further improve the detection rate of PSMA-based imaging. To achieve this, we tested dutasteride (Duta)-generally used for treatment of benign prostatic enlargement-and lovastatin (Lova)-a compound used to reduce blood lipid concentrations. We aimed to compare the individual effects of Duta and Lova on cell proliferation as well as PSMA expression. In addition, we tested if a combination treatment using lower concentrations of Duta and Lova can further induce PSMA expression. Our results show that a treatment with ≤1 μM Duta and ≥1 μM Lova lead to a significant upregulation of whole and cell surface PSMA expression in LNCaP, C4-2 and VCaP cells. Lower concentrations of Duta and Lova in combination (0.5 μM Duta + 0.5 μM Lova or 0.5 μM Duta + 1 μM Lova) were further capable of enhancing PSMA protein expression compared to a single compound treatment using higher concentrations in all tested cell lines (LNCaP, C4-2 and VCaP).
Collapse
Affiliation(s)
- Aleksandar Kuzmanov
- Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, University of Zurich, 8091 Zurich, Switzerland
| | - Souzan Salemi
- Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, University of Zurich, 8091 Zurich, Switzerland
| | - Florian A. Schmid
- Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, University of Zurich, 8091 Zurich, Switzerland
| | - Irene A. Burger
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
- Department of Nuclear Medicine, Baden Cantonal Hospital, 5404 Baden, Switzerland
| | - Daniel Eberli
- Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, University of Zurich, 8091 Zurich, Switzerland
| | - Benedikt Kranzbühler
- Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
23
|
Arese M, Mahmoudian M, Bussolino F. RNA aptamer-mediated gene therapy of prostate cancer: lessons from the past and future directions. Expert Opin Drug Deliv 2023; 20:1609-1621. [PMID: 38058168 DOI: 10.1080/17425247.2023.2292691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/04/2023] [Indexed: 12/08/2023]
Abstract
INTRODUCTION Prostate cancer (PCa) is one of the most prevalent cancers in the world, and the fifth cause of death from cancer in men. Among the non-surgical treatments for PCa, gene therapy strategies are in the early stages of development and recent clinical trials have provided new insights suggesting promising future. AREAS COVERED Recently, the creation of targeted gene delivery systems, based on specific PCa cell surface markers, has been viewed as a viable therapeutic approach. Prostate-specific membrane antigen (PSMA) is vastly expressed in nearly all prostate malignancies, and the intensity of expression increases with tumor aggressiveness, androgen independence, and metastasis. RNA aptamers are short and single-stranded oligonucleotides, which selectively bind to a specific ligand on the surface of the cells, which makes them fascinating small molecules for target delivery of therapeutics. PSMA-selective RNA aptamers represent great potential for developing targeted-gene delivery tools for PCa. EXPERT OPINION This review provides a thorough horizon for the researchers interested in developing targeted gene delivery systems for PCa via PSMA RNA aptamers. In addition, we provided general information about different prospects of RNA aptamers including discovery approaches, stability, safety, and pharmacokinetics.
Collapse
Affiliation(s)
- Marco Arese
- Department of Oncology, University of Torino, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Mohammad Mahmoudian
- Department of Oncology, University of Torino, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Federico Bussolino
- Department of Oncology, University of Torino, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| |
Collapse
|
24
|
Wang J, Ben-David R, Mehrazin R, Yang W, Tewari AK, Kyprianou N. Novel signatures of prostate cancer progression and therapeutic resistance. Expert Opin Ther Targets 2023; 27:1195-1206. [PMID: 38108262 DOI: 10.1080/14728222.2023.2293757] [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: 08/31/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
INTRODUCTION The extensive heterogeneity of prostate cancer (PCa) and multilayered complexity of progression to castration-resistant prostate cancer (CRPC) have contributed to the challenges of accurately monitoring advanced disease. Profiling of the tumor microenvironment with large-scale transcriptomic studies have identified gene signatures that predict biochemical recurrence, lymph node invasion, metastases, and development of therapeutic resistance through critical determinants driving CRPC. AREAS COVERED This review encompasses understanding of the role of different molecular determinants of PCa progression to lethal disease including the phenotypic dynamic of cell plasticity, EMT-MET interconversion, and signaling-pathways driving PCa cells to advance and metastasize. The value of liquid biopsies encompassing circulating tumor cells and extracellular vesicles to detect disease progression and emergence of therapeutic resistance in patients progressing to lethal disease is discussed. Relevant literature was added from PubMed portal. EXPERT OPINION Despite progress in the tumor-targeted therapeutics and biomarker discovery, distant metastasis and therapeutic resistance remain the major cause of mortality in patients with advanced CRPC. No single signature can encompass the tremendous phenotypic and genomic heterogeneity of PCa, but rather multi-threaded omics-derived and phenotypic markers tailored and validated into a multimodal signature.
Collapse
Affiliation(s)
- Jason Wang
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Reuben Ben-David
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Reza Mehrazin
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wei Yang
- Department of Pathology, Stony Brook University, New York, NY, USA
| | - Ashutosh K Tewari
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Natasha Kyprianou
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology & Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
25
|
Alessandrini L, Astolfi L, Daloiso A, Sbaraglia M, Mondello T, Zanoletti E, Franz L, Marioni G. Diagnostic, Prognostic, and Therapeutic Role for Angiogenesis Markers in Head and Neck Squamous Cell Carcinoma: A Narrative Review. Int J Mol Sci 2023; 24:10733. [PMID: 37445908 DOI: 10.3390/ijms241310733] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/14/2023] [Accepted: 06/25/2023] [Indexed: 07/15/2023] Open
Abstract
Despite refinements to diagnostic and therapeutic approaches over the last two decades, the outcome of patients with head and neck squamous cell carcinoma (HNSCC) has not shown substantial improvements, especially regarding those with advanced-stage disease. Angiogenesis is believed to be a turning point in the development of solid tumors, being a premise for mass growth and potential distant dissemination. Cancer-induced angiogenesis is a result of increased expression of angiogenic factors, decreased expression of anti-angiogenic factors, or a combination of both. The assessment of angiogenesis has also emerged as a potentially useful biological prognostic and predictive factor in HNSCC. The aim of this review is to assess the level of current knowledge on the neo-angiogenesis markers involved in the biology, behavior, and prognosis of HNSCC. A search (between 1 January 2012 and 10 October 2022) was run in PubMed, Scopus, and Web of Science electronic databases. After full-text screening and application of inclusion/exclusion criteria, 84 articles are included. The current knowledge and debate on angiogenesis in HNSCC presented in the eligible articles are stratified as follows: (i) diagnostic markers; (ii) prognostic markers; (iii) predictive markers; and (iv) markers with a potential therapeutic role. Angiogenesis is a biological and pathological indicator of malignancies progression and has negative implications in prognosis of some solid tumors; several signals capable of tripping the "angiogenic switch" have also been identified in HNSCC. Although several studies suggested that antiangiogenic agents might be a valuable adjunct to conventional chemo-radiation of HNSCC, their long-term therapeutic value remains uncertain. Further investigations are required on combinations of antiangiogenic agents with conventional chemotherapeutic ones, immunotherapeutic and molecularly targeted agents in HNSCC. Additional data are necessary to pinpoint which patients could benefit most from these treatments.
Collapse
Affiliation(s)
- Lara Alessandrini
- Surgical Pathology and Cytopathology Unit, Department of Medicine (DIMED), University of Padova, 35100 Padova, Italy
| | - Laura Astolfi
- Bioacustic Research Laboratory, Department of Neuroscience (DNS), University of Padova, 35100 Padova, Italy
| | - Antonio Daloiso
- Otolaryngology Section, Department of Neuroscience (DNS), University of Padova, 35100 Padova, Italy
| | - Marta Sbaraglia
- Surgical Pathology and Cytopathology Unit, Department of Medicine (DIMED), University of Padova, 35100 Padova, Italy
| | - Tiziana Mondello
- Otolaryngology Section, Department of Neuroscience (DNS), University of Padova, 35100 Padova, Italy
| | - Elisabetta Zanoletti
- Otolaryngology Section, Department of Neuroscience (DNS), University of Padova, 35100 Padova, Italy
| | - Leonardo Franz
- Otolaryngology Section, Department of Neuroscience (DNS), University of Padova, 35100 Padova, Italy
- Phoniatrics and Audiology Unit, Department of Neuroscience (DNS), University of Padova, 31100 Treviso, Italy
- Artificial Intelligence in Medicine and Innovation in Clinical Research and Methodology (PhD Program), Department of Clinical and Experimental Sciences, University of Brescia, 25100 Brescia, Italy
| | - Gino Marioni
- Phoniatrics and Audiology Unit, Department of Neuroscience (DNS), University of Padova, 31100 Treviso, Italy
| |
Collapse
|
26
|
Hu Y, Wang P, Dai W. Heterogeneous uptake of 18F-FDG and 18F-PSMA-1007 PET/CT in lung cancer and lymph node metastasis. BMC Pulm Med 2023; 23:73. [PMID: 36882747 PMCID: PMC9993722 DOI: 10.1186/s12890-023-02377-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/02/2023] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND PSMA PET/CT has shown excellent results in imaging of prostate cancer. However, some nonprostatic malignancies can also demonstrate 18 F-PSMA uptake, including primary lung cancer. 18 F-FDG PET/CT is widely employed in initial staging, response to therapy and follow-up assessment for lung cancer. Here we present an interesting case report on the different patterns of PSMA and FDG uptake between primary lung cancer and metastatic intrathoracic lymph node metastases in a patient with concurrent metastatic prostate cancer. CASE PRESENTATION A 70-year-old male underwent 18 F-FDG PET/CT and 18 F-PSMA-1007 PET/CT imaging due to suspicion primary lung cancer and prostate cancer. The patient eventually was diagnosed with non-small cell lung cancer (NSCLC) with mediastinal lymph node metastases and prostate cancer with left iliac lymph node and multiple bone metastases. Interestingly, our imaging revealed different patterns of tumor uptake detected on 18 F-FDG and 18 F-PSMA-1007 PET/CT in primary lung cancer and lymph node metastases. The primary lung lesion showed intense FDG uptake, and mild uptake with 18 F-PSMA-1007. Whereas the mediastinal lymph node metastases showed both intense FDG and PSMA uptake. The prostate lesion, left iliac lymph node, and multiple bone lesions showed significant PSMA uptake and negative FDG uptake. CONCLUSION In this case, there was a homogeneity of 18 F-FDG intense uptake between LC and metastatic lymph nodes, but a heterogeneity in 18 F-PSMA-1007 uptake. It illustrated that these molecular probes reflect the diversity of tumor microenvironments, which may help us understand the differences of the tumor response to treatment.
Collapse
Affiliation(s)
- Yuan Hu
- Department of Nuclear Medicine, The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443003, Hubei, China
| | - Peng Wang
- Department of Nuclear Medicine, The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443003, Hubei, China
| | - Wenli Dai
- Department of Nuclear Medicine, The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443003, Hubei, China.
| |
Collapse
|
27
|
Advances in Molecular Regulation of Prostate Cancer Cells by Top Natural Products of Malaysia. Curr Issues Mol Biol 2023; 45:1536-1567. [PMID: 36826044 PMCID: PMC9954984 DOI: 10.3390/cimb45020099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Prostate cancer (PCa) remains both a global health burden and a scientific challenge. We present a review of the molecular targets driving current drug discovery to fight this disease. Moreover, the preventable nature of most PCa cases represents an opportunity for phytochemicals as chemopreventive when adequately integrated into nutritional interventions. With a renovated interest in natural remedies as a commodity and their essential role in cancer drug discovery, Malaysia is looking towards capitalizing on its mega biodiversity, which includes the oldest rainforest in the world and an estimated 1200 medicinal plants. We here explore whether the list of top Malay plants prioritized by the Malaysian government may fulfill the potential of becoming newer, sustainable sources of prostate cancer chemotherapy. These include Andrographis paniculate, Centella asiatica, Clinacanthus nutans, Eurycoma longifolia, Ficus deltoidea, Hibiscus sabdariffa, Marantodes pumilum (syn. Labisia pumila), Morinda citrifolia, Orthosiphon aristatus, and Phyllanthus niruri. Our review highlights the importance of resistance factors such as Smac/DIABLO in cancer progression, the role of the CXCL12/CXCR4 axis in cancer metastasis, and the regulation of PCa cells by some promising terpenes (andrographolide, Asiatic acid, rosmarinic acid), flavonoids (isovitexin, gossypin, sinensetin), and alkylresorcinols (labisiaquinones) among others.
Collapse
|
28
|
The Potential of PSMA as a Vascular Target in TNBC. Cells 2023; 12:cells12040551. [PMID: 36831218 PMCID: PMC9954547 DOI: 10.3390/cells12040551] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Recent studies proving prostate-specific membrane antigen (PSMA) expression on triple-negative breast cancer (TNBC) cells and adjacent endothelial cells suggest PSMA as a promising target for therapy of until now not-targetable cancer entities. In this study, PSMA and its isoform expression were analyzed in different TNBC cells, breast cancer stem cells (BCSCs), and tumor-associated endothelial cells. PSMA expression was detected in 91% of the investigated TNBC cell lines. The PSMA splice isoforms were predominantly found in the BCSCs. Tumor-conditioned media from two TNBC cell lines, BT-20 (high full-length PSMA expression, PSMAΔ18 expression) and Hs578T (low full-length PSMA expression, no isoform expression), showed significant pro-angiogenic effect with induction of tube formation in endothelial cells. All TNBC cell lines induced PSMA expression in human umbilical vein endothelial cells (HUVEC). Significant uptake of radiolabeled ligand [68Ga]Ga-PSMA was detected in BCSC1 (4.2%), corresponding to the high PSMA expression. Moreover, hypoxic conditions increased the uptake of radiolabeled ligand [177Lu]Lu-PSMA in MDA-MB-231 (0.4% vs. 3.4%, under hypoxia and normoxia, respectively) and MCF-10A (0.3% vs. 3.0%, under normoxia and hypoxia, respectively) significantly (p < 0.001). [177Lu]Lu-PSMA-induced apoptosis rates were highest in BT-20 and MDA-MB-231 associated endothelial cells. Together, these findings demonstrate the potential of PSMA-targeted therapy in TNBC.
Collapse
|
29
|
Pei Y, Liu C, Feng M, Li L, Zhou C, Chen L, Hu X, Song S, Cao Y, Gao Y. The clinical application of 68Ga-PSMA PET/CT and regulating mechanism of PSMA expression in patients with brain metastases of lung cancer. Transl Oncol 2023; 28:101616. [PMID: 36621073 PMCID: PMC9850174 DOI: 10.1016/j.tranon.2023.101616] [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: 07/21/2022] [Revised: 10/18/2022] [Accepted: 01/02/2023] [Indexed: 01/08/2023] Open
Abstract
Brain metastases (BMs) of lung cancer are common malignant intracranial tumours associated with severe neurological symptoms and an abysmal prognosis. Prostate-specific membrane antigen (PSMA) has been reported to express significantly in a variety of solid tumours. However, the clinical applications of 68Ga-PSMA PET/CT and the mechanism of PSMA expression in patients with BMs of lung cancer have rarely been reported. Experiments with 68Ga-PSMA PET/CT and immunohistochemical staining were conducted to evaluate the expression of PSMA from seven patients with BMs of lung cancer who accepted surgical treatment in Fudan University Shanghai Cancer Center between October 2020 and October 2021. The mechanism of PSMA expression in BMs of lung cancer was explored by using single-cell RNA sequencing. The median maximum standardized uptake value (SUVmax) in BMs was higher than that in primary lung cancer (8.6 ± 2.8 vs. 3.6 ± 1.3, P < 0.01). The mean SUVmax in BMs was 1.76-fold higher than that in the liver, which indicated the potential of PSMA radioligand therapy (PSMA-RLT) for BMs. BMs showed intense PSMA staining, while normal lung tissue had no PSMA staining and there was only faint primary lung cancer staining by immunohistochemistry (IHC). Single-cell RNA sequencing (scRNA-seq) analysis found that PSMA was mainly expressed in oligodendrocytes of BMs, whereas it was expressed at lower levels in solid cells of lung cancer. PSMA expression in oligodendrocytes might be regulated by the factors ATF3 and NR4A1, which were associated with ER stress.
Collapse
Affiliation(s)
- Yuchen Pei
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Chang Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Mingtao Feng
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Liangdong Li
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Changshuai Zhou
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lei Chen
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xin Hu
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yiqun Cao
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yang Gao
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, China,Corresponding author.
| |
Collapse
|
30
|
Lindemann M, Oteiza A, Martin-Armas M, Guttormsen Y, Moldes-Anaya A, Berzaghi R, Bogsrud TV, Bach-Gansmo T, Sundset R, Kranz M. Glioblastoma PET/MRI: kinetic investigation of [ 18F]rhPSMA-7.3, [ 18F]FET and [ 18F]fluciclovine in an orthotopic mouse model of cancer. Eur J Nucl Med Mol Imaging 2023; 50:1183-1194. [PMID: 36416908 PMCID: PMC9931868 DOI: 10.1007/s00259-022-06040-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/11/2022] [Indexed: 11/24/2022]
Abstract
PURPOSE Glioblastoma multiforme (GBM) is the most common glioma and standard therapies can only slightly prolong the survival. Neo-vascularization is a potential target to image tumor microenvironment, as it defines its brain invasion. We investigate [18F]rhPSMA-7.3 with PET/MRI for quantitative imaging of neo-vascularization in GBM bearing mice and human tumor tissue and compare it to [18F]FET and [18F]fluciclovine using PET pharmacokinetic modeling (PKM). METHODS [18F]rhPSMA-7.3, [18F]FET, and [18F]fluciclovine were i.v. injected with 10.5 ± 3.1 MBq, 8.0 ± 2.2 MBq, 11.5 ± 1.9 MBq (n = 28, GL261-luc2) and up to 90 min PET/MR imaged 21/28 days after surgery. Regions of interest were delineated on T2-weighted MRI for (i) tumor, (ii) brain, and (iii) the inferior vena cava. Time-activity curves were expressed as SUV mean, SUVR and PKM performed using 1-/2-tissue-compartment models (1TCM, 2TCM), Patlak and Logan analysis (LA). Immunofluorescent staining (IFS), western blotting, and autoradiography of tumor tissue were performed for result validation. RESULTS [18F]rhPSMA-7.3 showed a tumor uptake with a tumor-to-background-ratio (TBR) = 2.1-2.5, in 15-60 min. PKM (2TCM) confirmed higher K1 (0.34/0.08, p = 0.0012) and volume of distribution VT (0.24/0.1, p = 0.0017) in the tumor region compared to the brain. Linearity in LA and similar k3 = 0.6 and k4 = 0.47 (2TCM, tumor, p = ns) indicated reversible binding. K1, an indicator for vascularization, increased (0.1/0.34, 21 to 28 days, p < 0.005). IFS confirmed co-expression of PSMA and tumor vascularization. [18F]fluciclovine showed higher TBR (2.5/1.8, p < 0.001, 60 min) and VS (1.3/0.7, p < 0.05, tumor) compared to [18F]FET and LA indicated reversible binding. VT increased (p < 0.001, tumor, 21 to 28 days) for [18F]FET (0.5-1.4) and [18F]fluciclovine (0.84-1.5). CONCLUSION [18F]rhPSMA-7.3 showed to be a potential candidate to investigate the tumor microenvironment of GBM. Following PKM, this uptake was associated with tumor vascularization. In contrast to what is known from PSMA-PET in prostate cancer, reversible binding was found for [18F]rhPSMA-7.3 in GBM, contradicting cellular trapping. Finally, [18F]fluciclovine was superior to [18F]FET rendering it more suitable for PET imaging of GBM.
Collapse
Affiliation(s)
- Marcel Lindemann
- PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway
- Nuclear Medicine and Radiation Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Ana Oteiza
- PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway
- Nuclear Medicine and Radiation Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Montserrat Martin-Armas
- PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway
- Nuclear Medicine and Radiation Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Yngve Guttormsen
- PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway
- Nuclear Medicine and Radiation Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Angel Moldes-Anaya
- PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway
- Nuclear Medicine and Radiation Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Rodrigo Berzaghi
- Nuclear Medicine and Radiation Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Trond Velde Bogsrud
- PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway
- PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Tore Bach-Gansmo
- PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway
| | - Rune Sundset
- PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway
- Nuclear Medicine and Radiation Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Mathias Kranz
- PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway.
- Nuclear Medicine and Radiation Biology, UiT The Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
31
|
Zhang J, Lu T, Lu S, Ma S, Han D, Zhang K, Xu C, Liu S, Gan L, Wu X, Yang F, Wen W, Qin W. Single-cell analysis of multiple cancer types reveals differences in endothelial cells between tumors and normal tissues. Comput Struct Biotechnol J 2022; 21:665-676. [PMID: 36659929 PMCID: PMC9826920 DOI: 10.1016/j.csbj.2022.12.049] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 12/31/2022] Open
Abstract
Endothelial cells (ECs) play an important role in tumor progression. Currently, the main target of anti-angiogenic therapy is the vascular endothelial growth factor (VEGF) pathway. Some patients do benefit from anti-VEGF/VEGFR therapy; however, a large number of patients do not have response or acquire drug resistance after treatment. Moreover, anti-VEGF/VEGFR therapy may lead to nephrotoxicity and cardiovascular-related side effects due to its action on normal ECs. Therefore, it is necessary to identify targets that are specific to tumor ECs and could be applied to various cancer types. We integrated single-cell RNA sequencing data from six cancer types and constructed a multi-cancer EC atlas to decode the characteristic of tumor ECs. We found that tip-like ECs mainly exist in tumor tissues but barely exist in normal tissues. Tip-like ECs are involved in the promotion of tumor angiogenesis and inhibition on anti-tumor immune responses. Moreover, tumor cells, myeloid cells, and pericytes are the main sources of pro-angiogenic factors. High proportion of tip-like ECs is associated with poor prognosis in multiple cancer types. We also identified that prostate-specific membrane antigen (PSMA) is a specific marker for tip-like ECs in all the cancer types we studied. In summary, we demonstrate that tip-like ECs are the main differential EC subcluster between tumors and normal tissues. Tip-like ECs may promote tumor progression through promoting angiogenesis while inhibiting anti-tumor immune responses. PSMA was a specific marker for tip-like ECs, which could be used as a potential target for the diagnosis and treatment of non-prostate cancers.
Collapse
Key Words
- BRCA, Breast invasive carcinoma
- CESC, Cervical squamous cell carcinoma and endocervical adenocarcinoma
- CRC, Colorectal cancer
- ECs, Endothelial cells
- Endothelial cells
- GC, Gastric cancer
- HNSC, Head and Neck squamous cell carcinoma
- KICH, Kidney chromophobe
- KIRC, Kidney renal clear cell carcinoma
- KIRP, Kidney renal papillary cell carcinoma
- LC, Lung cancer
- LIHC, Liver hepatocellular carcinoma
- LUAD, Lung adenocarcinoma
- LUSC, Lung squamous cell carcinoma
- OV, Ovarian serous cystadenocarcinoma
- OVC, Ovarian cancer
- PAAD, Pancreatic adenocarcinoma
- PDAC, Pancreatic ductal adenocarcinoma
- PRAD, Prostate adenocarcinoma
- PSMA, Prostate-specific membrane antigen
- RCC, Renal cell carcinoma
- READ, Rectum adenocarcinoma
- STAD, Stomach adenocarcinoma
- Single-cell RNA sequencing
- TME, Tumor microenvironment
- Tumor microenvironment
- VEGF, Vascular endothelial growth factor
- scRNA-seq, Single-cell RNA sequencing
Collapse
Affiliation(s)
- Jiayu Zhang
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Tong Lu
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Shiqi Lu
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Shuaijun Ma
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Donghui Han
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Keying Zhang
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Chao Xu
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Shaojie Liu
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Lunbiao Gan
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Xinjie Wu
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Fa Yang
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi’an, China,Correspondence to: Department of Urology, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, China.
| | - Weihong Wen
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China,Correspondence to: Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, China.
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi’an, China,Correspondence to: Department of Urology, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, China.
| |
Collapse
|
32
|
Godê KKDS, Mourato FA, Sales AFDF, de Almeida Filho PJ, Brandão SCS, Wichert-Ana L. Thyroid incidentalomas in PSMA PET/CT: a systematic review and meta-analysis. Clin Transl Imaging 2022. [DOI: 10.1007/s40336-022-00537-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
33
|
Ameen M, Sundaram L, Shen M, Banerjee A, Kundu S, Nair S, Shcherbina A, Gu M, Wilson KD, Varadarajan A, Vadgama N, Balsubramani A, Wu JC, Engreitz JM, Farh K, Karakikes I, Wang KC, Quertermous T, Greenleaf WJ, Kundaje A. Integrative single-cell analysis of cardiogenesis identifies developmental trajectories and non-coding mutations in congenital heart disease. Cell 2022; 185:4937-4953.e23. [PMID: 36563664 PMCID: PMC10122433 DOI: 10.1016/j.cell.2022.11.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 09/13/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022]
Abstract
To define the multi-cellular epigenomic and transcriptional landscape of cardiac cellular development, we generated single-cell chromatin accessibility maps of human fetal heart tissues. We identified eight major differentiation trajectories involving primary cardiac cell types, each associated with dynamic transcription factor (TF) activity signatures. We contrasted regulatory landscapes of iPSC-derived cardiac cell types and their in vivo counterparts, which enabled optimization of in vitro differentiation of epicardial cells. Further, we interpreted sequence based deep learning models of cell-type-resolved chromatin accessibility profiles to decipher underlying TF motif lexicons. De novo mutations predicted to affect chromatin accessibility in arterial endothelium were enriched in congenital heart disease (CHD) cases vs. controls. In vitro studies in iPSCs validated the functional impact of identified variation on the predicted developmental cell types. This work thus defines the cell-type-resolved cis-regulatory sequence determinants of heart development and identifies disruption of cell type-specific regulatory elements in CHD.
Collapse
Affiliation(s)
- Mohamed Ameen
- Department of Cancer Biology, Stanford University, Stanford, CA, USA; Illumina Artificial Intelligence Laboratory, Illumina Inc, Foster City, CA, USA
| | - Laksshman Sundaram
- Department of Computer Science, Stanford University, Stanford, CA, USA; Illumina Artificial Intelligence Laboratory, Illumina Inc, Foster City, CA, USA
| | - Mengcheng Shen
- Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Abhimanyu Banerjee
- Illumina Artificial Intelligence Laboratory, Illumina Inc, Foster City, CA, USA; Department of Physics, Stanford University, Stanford, CA, USA
| | - Soumya Kundu
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Surag Nair
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Anna Shcherbina
- Department of Biomedical Informatics, Stanford University, Stanford, CA, USA
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine, CuSTOM, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | - Avyay Varadarajan
- Department of Computer Science, California Institute of Technology, Pasadena, CA, USA
| | - Nirmal Vadgama
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | | | - Joseph C Wu
- Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | | | - Kyle Farh
- Illumina Artificial Intelligence Laboratory, Illumina Inc, Foster City, CA, USA
| | - Ioannis Karakikes
- Cardiovascular Institute, Stanford University, Stanford, CA, USA; Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA.
| | - Kevin C Wang
- Department of Cancer Biology, Stanford University, Stanford, CA, USA; Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA; Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA, USA.
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - William J Greenleaf
- Department of Genetics, Stanford University, Stanford, CA, USA; Department of Applied Physics, Stanford University, Stanford, CA, USA.
| | - Anshul Kundaje
- Department of Computer Science, Stanford University, Stanford, CA, USA; Department of Genetics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
34
|
Kmeid M, Park YN, Chung T, Lukose G, Sullivan L, Brar R, Lee H. PSMA Immunohistochemistry in Hepatic Neoplasms: A Promising Diagnostic Marker With Potential Theranostic Applications. Am J Surg Pathol 2022; 46:1688-1699. [PMID: 36190927 DOI: 10.1097/pas.0000000000001971] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Accurate classification of well-differentiated hepatocellular neoplasms can be challenging especially in core biopsies. Prostate-specific membrane antigen (PSMA) has been shown to highlight tumor-associated neovasculature in many nonprostatic solid tumors including hepatocellular carcinoma (HCC). Archived 164 hepatectomies and explants with 68 HCCs, 31 hepatocellular adenoma (HA), 24 dysplastic nodules (DN), and 42 metastases were retrieved, and pathologic parameters were evaluated. Sensitivity, specificity, accuracy, positive, and negative predictive values for correct diagnosis of HCC were calculated for PSMA and CD34 immunostains in tissue sections and HCC tissue microarrays. PSMA positivity was defined as capillarized sinusoidal/tumor-associated vessel staining involving ≥5% of the tumor area. In all, 55/68 (80.9%) HCC and 37/42 (88.1%) of liver metastasis were PSMA positive. PSMA was negative in HA, DN, and background liver (100% specificity). CD34 had a 98.5% sensitivity but a 65.5% specificity in identifying HCC. PSMA sensitivity remained high in the HCC tissue microarray (89.7%). PSMA was more accurate than CD34 (95.5% vs. 69.7%) in distinguishing grade 1 HCC from HA and high-grade DN while retaining high sensitivity (80%). The degree of PSMA positivity in HCC was greater in older, male, and human immunodeficiency virus patients ( P <0.05). No associations were found between PSMA staining and other tumor parameters ( P >0.05). PSMA is a marker of neoangiogenesis with increased expression in both primary and metastatic hepatic malignancies. Neovascular PSMA expression is more specific and accurate than CD34 for differentiating HCC from benign and precursor hepatic lesions. Diagnostic and therapeutic utility of PSMA radioligands in malignant liver neoplasms warrant further clinical investigations.
Collapse
Affiliation(s)
- Michel Kmeid
- Department of Pathology, Albany Medical Center, Albany
| | | | - Taek Chung
- Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Georgi Lukose
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Luz Sullivan
- Department of Pathology, Albany Medical Center, Albany
| | - Rupinder Brar
- Department of Pathology, Albany Medical Center, Albany
| | - Hwajeong Lee
- Department of Pathology, Albany Medical Center, Albany
| |
Collapse
|
35
|
Digklia A, Boughdad S, Homicsko K, Dromain C, Trimech M, Dolcan A, Peters S, Prior J, Schaefer N. First communication on the efficacy of combined <sup>177</sup>Lutetium-PSMA with immunotherapy outside prostate cancer. J Immunother Cancer 2022; 10:jitc-2022-005383. [PMID: 36288828 PMCID: PMC9615971 DOI: 10.1136/jitc-2022-005383] [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] [Accepted: 09/12/2022] [Indexed: 11/23/2022] Open
Abstract
Prostate-specific membrane antigen (PSMA)-targeted radioligand therapy is a validated treatment option for patients with advanced prostate cancer. Although PSMA expression is not limited to prostate tissue, little is known about its relevance to other types of cancer. Here, we present a case report of a patient with uterine leiomyosarcoma that is progressing while on immunotherapy and treated with <sup>177</sup>Lu-PSMA radionuclide therapy. We report for the first time that <sup>177</sup>Lu-PSMA radionuclide therapy combined with immunotherapy outside of prostate cancer. We did observe post-treatment reduction of tumor growth rate, although we did not notice disease response based on RECIST criteria. We suggest that <sup>177</sup>Lu-PSMA treatment especially combined with immunotherapy may be an option for patients with cancer without other therapeutic options. Insights: <sup>177</sup>Lu-PSMA radionuclide therapy should be considered for any tumor stained positive for PSMA.
Collapse
Affiliation(s)
- Antonia Digklia
- Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland,Sarcoma Center, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland,University of Lausanne, Lausanne, Switzerland
| | - Sarah Boughdad
- Nuclear Medicine and Molecular Imaging, University of Lausanne, Lausanne, Switzerland
| | - Krisztian Homicsko
- Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland,University of Lausanne, Lausanne, Switzerland,Precision Oncology Center, Lausanne University Hospital, Lausanne, Switzerland
| | - Clarisse Dromain
- University of Lausanne, Lausanne, Switzerland,Radiology and Interventional Radiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Mounir Trimech
- University of Lausanne, Lausanne, Switzerland,Pathology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Ana Dolcan
- Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland,Sarcoma Center, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Solange Peters
- Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland,University of Lausanne, Lausanne, Switzerland
| | - John Prior
- University of Lausanne, Lausanne, Switzerland,Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Niklaus Schaefer
- Sarcoma Center, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland,University of Lausanne, Lausanne, Switzerland,Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
36
|
Marinescu IM, Rogg M, Spohn S, von Büren M, Kamps M, Jilg CA, Fountzila E, Papadopoulou K, Ceci L, Bettermann A, Ruf J, Benndorf M, Adebahr S, Zips D, Grosu AL, Schell C, Zamboglou C. Ex vivo γH2AX assay for tumor radiosensitivity in primary prostate cancer patients and correlation with clinical parameters. Radiat Oncol 2022; 17:163. [PMID: 36199143 PMCID: PMC9533509 DOI: 10.1186/s13014-022-02131-1] [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: 08/22/2022] [Accepted: 09/13/2022] [Indexed: 11/15/2022] Open
Abstract
Backround Accurate surrogate parameters for radio resistance are warranted for individualized radiotherapy (RT) concepts in prostate cancer (PCa). The purpose of this study was to assess intertumoral heterogeneity in terms of radio resistance using an ex-vivo γH2AX assay after irradiation of prostate biopsy cores and to investigate its correlation with clinical features of respective patients as well as imaging and genomic features of tumor areas.
Methods Twenty one patients with histologically-proven PCa and pre-therapeutic multiparametric resonance imaging and prostate-specific membrane antigen positron emission tomography were included in the study. Biopsy cores were collected from 26 PCa foci. Residual γH2AX foci were counted 24 h after ex-vivo irradiation (with 0 and 4 Gy) of biopsy specimen and served as a surrogate for radio resistance. Clinical, genomic (next generation sequencing) and imaging features were collected and their association with the radio resistance was studied. Results In total 18 PCa lesions from 16 patients were included in the final analysis. The median γH2AX foci value per PCa lesion was 3.12. According to this, the patients were divided into two groups (radio sensitive vs. radio resistant) with significant differences in foci number (p < 0.0001). The patients in the radio sensitive group had significantly higher prostate specific antigen serum concentration (p = 0.015), tumor areas in the radio sensitive group had higher SUV (standardized uptake values in PSMA PET)-max and -mean values (p = 0.0037, p = 0.028) and lower ADC (apparent diffusion coefficient-mean values, p = 0.049). All later parameters had significant (p < 0.05) correlations in Pearson’s test. One patient in the radio sensitive group displayed a previously not reported loss of function frameshift mutation in the NBN gene (c.654_658delAAAAC) that introduces a premature termination codon and results in a truncated protein. Conclusion In this pilot study, significant differences in intertumoral radio resistance were observed and clinical as well as imaging parameters may be applied for their prediction. After further prospective validation in larger patient cohorts these finding may lead to individual RT dose prescription for PCa patients in the future.
Collapse
Affiliation(s)
- Ioana M Marinescu
- Department of Radiation Oncology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany. .,German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany.
| | - Manuel Rogg
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Simon Spohn
- Department of Radiation Oncology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany
| | - Moritz von Büren
- Department of Urology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Marius Kamps
- Department of Urology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Cordula A Jilg
- Department of Urology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Elena Fountzila
- Second Department of Medical Oncology, Euromedica General Clinic of Thessaloniki, Thessaloniki, Greece.,Greece and European University Cyprus, Engomi, Cyprus
| | - Kyriaki Papadopoulou
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Lara Ceci
- Department of Radiation Oncology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany
| | - Alisa Bettermann
- Department of Radiation Oncology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany
| | - Juri Ruf
- Department of Nuclear Medicine, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Matthias Benndorf
- Department of Radiology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Sonja Adebahr
- Department of Radiation Oncology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany
| | - Daniel Zips
- Medical Faculty and University Hospital, Radiation Oncology, Eberhard Karls University Tübingen, Tübingen, Germany.,German Cancer Consortium (DKTK), Partner Site Tübingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anca L Grosu
- Department of Radiation Oncology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany
| | - Christoph Schell
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany.,Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Tumorbank Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, University of Freiburg, Freiburg, Germany.,German Oncology Center, European University Cyprus, Limassol, Cyprus
| |
Collapse
|
37
|
Gori SS, Thomas AG, Pal A, Wiseman R, Ferraris DV, Gao RD, Wu Y, Alt J, Tsukamoto T, Slusher BS, Rais R. D-DOPA Is a Potent, Orally Bioavailable, Allosteric Inhibitor of Glutamate Carboxypeptidase II. Pharmaceutics 2022; 14:pharmaceutics14102018. [PMID: 36297453 PMCID: PMC9608075 DOI: 10.3390/pharmaceutics14102018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/14/2022] [Accepted: 09/19/2022] [Indexed: 12/02/2022] Open
Abstract
Glutamate carboxypeptidase-II (GCPII) is a zinc-dependent metalloenzyme implicated in numerous neurological disorders. The pharmacophoric requirements of active-site GCPII inhibitors makes them highly charged, manifesting poor pharmacokinetic (PK) properties. Herein, we describe the discovery and characterization of catechol-based inhibitors including L-DOPA, D-DOPA, and caffeic acid, with sub-micromolar potencies. Of these, D-DOPA emerged as the most promising compound, with good metabolic stability, and excellent PK properties. Orally administered D-DOPA yielded high plasma exposures (AUCplasma = 72.7 nmol·h/mL) and an absolute oral bioavailability of 47.7%. Unfortunately, D-DOPA brain exposures were low with AUCbrain = 2.42 nmol/g and AUCbrain/plasma ratio of 0.03. Given reports of isomeric inversion of D-DOPA to L-DOPA via D-amino acid oxidase (DAAO), we evaluated D-DOPA PK in combination with the DAAO inhibitor sodium benzoate and observed a >200% enhancement in both plasma and brain exposures (AUCplasma = 185 nmol·h/mL; AUCbrain = 5.48 nmol·h/g). Further, we demonstrated GCPII target engagement; orally administered D-DOPA with or without sodium benzoate caused significant inhibition of GCPII activity. Lastly, mode of inhibition studies revealed D-DOPA to be a noncompetitive, allosteric inhibitor of GCPII. To our knowledge, this is the first report of D-DOPA as a distinct scaffold for GCPII inhibition, laying the groundwork for future optimization to obtain clinically viable candidates.
Collapse
Affiliation(s)
- Sadakatali S. Gori
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ajit G. Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Arindom Pal
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Robyn Wiseman
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Dana V. Ferraris
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Run-duo Gao
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ying Wu
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Takashi Tsukamoto
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Barbara S. Slusher
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Correspondence: (B.S.S.); (R.R.)
| | - Rana Rais
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Correspondence: (B.S.S.); (R.R.)
| |
Collapse
|
38
|
Tariq A, McCart Reed AE, Morton A, Porten S, Vela I, Williams ED, Yaxley JW, Black PC, Roberts MJ. Urothelial Carcinoma and Prostate-specific Membrane Antigen: Cellular, Imaging, and Prognostic Implications. Eur Urol Focus 2022; 8:1256-1269. [PMID: 34429271 DOI: 10.1016/j.euf.2021.07.016] [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: 05/18/2021] [Revised: 07/17/2021] [Accepted: 07/29/2021] [Indexed: 12/16/2022]
Abstract
CONTEXT Staging, restaging, and surveillance of urothelial carcinoma (UC) is challenging due to suboptimal accuracy of standard of care imaging modalities. Prostate-specific membrane antigen (PSMA) imaging may serve to improve characterisation of UC. OBJECTIVE To appraise available literature regarding cellular, imaging, and prognostic implications of PSMA for UC. EVIDENCE ACQUISITION A systematic review was performed considering all available literature (including conference abstracts) published from 1990 to 2020 and reported according to Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines following registration in PROSPERO (CRD42020186744). All relevant texts relating to immunohistochemical analysis and PSMA-based imaging in UC were included and collated. Additionally, FOLH1 (gene encoding PSMA) expression according to The Cancer Genome Atlas (TCGA) database was analysed as well as according to consensus and TCGA molecular classification subtypes and subsequently compared with clinical outcomes. EVIDENCE SYNTHESIS PSMA expression across UC tumour tissue was heterogeneous (0-100%) but appeared to decrease with increased grade and stage. The TCGA analysis demonstrated loss of FOLH1 expression with increasing T stage (p = 0.0180) and N stage (p = 0.0269), and reduced FOLH1 expression was associated with worse disease-free survival. PSMA expression in UC neovasculature was variable but mostly increased (44-100%). Eleven reports of PSMA-based imaging for UC were identified, reporting on 18 patients. PSMA positron emission tomography (PET) imaging was positive in 17 out of 18 patients. The included literature review data were limited by mostly low-quality, retrospective studies. CONCLUSIONS Tissue PSMA, or FOLH1 expression, may inversely be associated with pathological and survival outcomes in localised UC. PSMA PET imaging may improve detection of metastatic disease and response to systemic therapy due to PSMA expression in neovasculature. Available evidence is limited; thus, larger, prospective studies are required to confirm early results and define populations that benefit most. PATIENT SUMMARY In this systematic review, we assess the potential role of prostate-specific membrane antigen in urothelial cancer. We found that its utility is in expression of blood vessels surrounding metastasis. We conclude that it may be beneficial in detecting metastasis and response to systemic therapies.
Collapse
Affiliation(s)
- Arsalan Tariq
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Amy E McCart Reed
- University of Queensland Centre for Clinical Research, Faculty of Medicine, Brisbane, Queensland, Australia
| | - Andrew Morton
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Sima Porten
- Department of Urology, University of California San Francisco, San Francisco, CA, USA
| | - Ian Vela
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia; Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia; Australian Prostate Cancer Research Centre-Queensland, Brisbane, Queensland, Australia; School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia; Translational Research Institute, Brisbane, Queensland, Australia
| | - Elizabeth D Williams
- Australian Prostate Cancer Research Centre-Queensland, Brisbane, Queensland, Australia; School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia; Translational Research Institute, Brisbane, Queensland, Australia
| | - John W Yaxley
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Peter C Black
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Matthew J Roberts
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; University of Queensland Centre for Clinical Research, Faculty of Medicine, Brisbane, Queensland, Australia; Department of Urology, Redcliffe Hospital, Brisbane, Queensland, Australia.
| |
Collapse
|
39
|
Rizzo A, Dall’Armellina S, Pizzuto DA, Perotti G, Zagaria L, Lanni V, Treglia G, Racca M, Annunziata S. PSMA Radioligand Uptake as a Biomarker of Neoangiogenesis in Solid Tumours: Diagnostic or Theragnostic Factor? Cancers (Basel) 2022; 14:4039. [PMID: 36011032 PMCID: PMC9406909 DOI: 10.3390/cancers14164039] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/10/2022] [Accepted: 08/17/2022] [Indexed: 01/10/2023] Open
Abstract
Due to its overexpression on the surface of prostate cancer cells, prostate-specific membrane antigen (PSMA) is a relatively novel effective target for molecular imaging and radioligand therapy (RLT) in prostate cancer. Recent studies reported that PSMA is expressed in the neovasculature of various types of cancer and regulates tumour cell invasion as well as tumour angiogenesis. Several authors explored the role of diagnostic and therapeutic PSMA radioligands in various malignancies. In this narrative review, we describe the current status of the literature on PSMA radioligands' application in solid tumours other than prostate cancer to explore their potential role as diagnostic or therapeutic agents, with particular regard to the relevance of PSMA radioligand uptake as neoangiogenetic biomarker. Hence, a comprehensive review of the literature was performed to find relevant articles on the applications of PSMA radioligands in non-prostate solid tumours. Data on the general, methodological and clinical aspects of all included studies were collected. Forty full-text papers were selected for final review, 8 of which explored PSMA radioligand PET/CT performances in gliomas, 3 in salivary gland malignancies, 6 in thyroid cancer, 2 in breast cancer, 16 in renal cell carcinoma and 5 in hepatocellular carcinoma. In the included studies, PSMA radioligand PET showed promising performance in patients with non-prostate solid tumours. Further studies are needed to better define its potential role in oncological patients management, especially in those undergoing antineoangiogenic therapies, and to assess the efficacy of PSMA-RLT in this clinical context.
Collapse
Affiliation(s)
- Alessio Rizzo
- Department of Nuclear Medicine, Candiolo Cancer Institute, FPO—IRCCS, 10060 Turin, Italy
| | - Sara Dall’Armellina
- Nuclear Medicine Unit, Department of Medical Sciences, AOU Città della Salute e della Scienza, University of Turin, 10134 Turin, Italy
| | - Daniele Antonio Pizzuto
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Germano Perotti
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Luca Zagaria
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Valerio Lanni
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giorgio Treglia
- Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6501 Bellinzona, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Manuela Racca
- Department of Nuclear Medicine, Candiolo Cancer Institute, FPO—IRCCS, 10060 Turin, Italy
| | - Salvatore Annunziata
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| |
Collapse
|
40
|
An S, Huang G, Liu J, Wei W. PSMA-targeted theranostics of solid tumors: applications beyond prostate cancers. Eur J Nucl Med Mol Imaging 2022; 49:3973-3976. [PMID: 35916921 DOI: 10.1007/s00259-022-05905-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Shuxian An
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
41
|
Sheehan B, Neeb A, Buroni L, Paschalis A, Riisnaes R, Gurel B, Gil V, Miranda S, Crespo M, Guo C, Jiménez Vacas J, Figueiredo I, Ferreira A, Welti J, Yuan W, Carreira S, Sharp A, de Bono J. Prostate-Specific Membrane Antigen Expression and Response to DNA Damaging Agents in Prostate Cancer. Clin Cancer Res 2022; 28:3104-3115. [PMID: 35552383 PMCID: PMC9365343 DOI: 10.1158/1078-0432.ccr-21-4531] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/15/2022] [Accepted: 05/09/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE Prostate-specific membrane antigen (PSMA) targeting therapies such as Lutetium-177 (177Lu)-PSMA-617 are affecting outcomes from metastatic castration-resistant prostate cancer (mCRPC). However, a significant subset of patients have prostate cancer cells lacking PSMA expression, raising concerns about treatment resistance attributable at least in part to heterogeneous PSMA expression. We have previously demonstrated an association between high PSMA expression and DNA damage repair defects in mCRPC biopsies and therefore hypothesized that DNA damage upregulates PSMA expression. EXPERIMENTAL DESIGN To test this relationship between PSMA and DNA damage we conducted a screen of 147 anticancer agents (NCI/NIH FDA-approved anticancer "Oncology Set") and treated tumor cells with repeated ionizing irradiation. RESULTS The topoisomerase-2 inhibitors, daunorubicin and mitoxantrone, were identified from the screen to upregulate PSMA protein expression in castration-resistant LNCaP95 cells; this result was validated in vitro in LNCaP, LNCaP95, and 22Rv1 cell lines and in vivo using an mCRPC patient-derived xenograft model CP286 identified to have heterogeneous PSMA expression. As double-strand DNA break induction by topoisomerase-2 inhibitors upregulated PSMA, we next studied the impact of ionizing radiation on PSMA expression; this also upregulated PSMA protein expression in a dose-dependent fashion. CONCLUSIONS The results presented herein are the first, to our knowledge, to demonstrate that PSMA is upregulated in response to double-strand DNA damage by anticancer treatment. These data support the study of rational combinations that maximize the antitumor activity of PSMA-targeted therapeutic strategies by upregulating PSMA.
Collapse
Affiliation(s)
| | - Antje Neeb
- The Institute of Cancer Research, London, UK
| | | | - Alec Paschalis
- The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, Sutton, UK
| | | | - Bora Gurel
- The Institute of Cancer Research, London, UK
| | | | | | | | - Christina Guo
- The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, Sutton, UK
| | | | | | | | - Jon Welti
- The Institute of Cancer Research, London, UK
| | - Wei Yuan
- The Institute of Cancer Research, London, UK
| | | | - Adam Sharp
- The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Johann de Bono
- The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, Sutton, UK
| |
Collapse
|
42
|
Thompson SM, Suman G, Torbenson MS, Chen ZE, Jondal DE, Patra A, Ehman EC, Andrews JC, Fleming CJ, Welch BT, Kurup AN, Roberts LR, Watt KD, Truty MJ, Cleary SP, Smoot RL, Heimbach JK, Tran NH, Mahipal A, Yin J, Zemla T, Wang C, Fogarty Z, Jacobson M, Kemp BJ, Venkatesh SK, Johnson GB, Woodrum DA, Goenka AH. PSMA as a Theranostic Target in Hepatocellular Carcinoma: Immunohistochemistry and 68 Ga-PSMA-11 PET Using Cyclotron-Produced 68 Ga. Hepatol Commun 2022; 6:1172-1185. [PMID: 34783177 PMCID: PMC9035563 DOI: 10.1002/hep4.1861] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 10/18/2021] [Accepted: 10/26/2021] [Indexed: 02/05/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA) is a validated target for molecular diagnostics and targeted radionuclide therapy. Our purpose was to evaluate PSMA expression in hepatocellular carcinoma (HCC), cholangiocarcinoma (CCA), and hepatic adenoma (HCA); investigate the genetic pathways in HCC associated with PSMA expression; and evaluate HCC detection rate with 68 Ga-PSMA-11 positron emission tomography (PET). In phase 1, PSMA immunohistochemistry (IHC) on HCC (n = 148), CCA (n = 111), and HCA (n = 78) was scored. In a subset (n = 30), messenger RNA (mRNA) data from the Cancer Genome Atlas HCC RNA sequencing were correlated with PSMA expression. In phase 2, 68 Ga-PSMA-11 PET was prospectively performed in patients with treatment-naïve HCC on a digital PET scanner using cyclotron-produced 68 Ga. Uptake was graded qualitatively and semi-quantitatively using standard metrics. On IHC, PSMA expression was significantly higher in HCC compared with CCA and HCA (P < 0.0001); 91% of HCCs (n = 134) expressed PSMA, which principally localized to tumor-associated neovasculature. Higher tumor grade was associated with PSMA expression (P = 0.012) but there was no association with tumor size (P = 0.14), fibrosis (P = 0.35), cirrhosis (P = 0.74), hepatitis B virus (P = 0.31), or hepatitis C virus (P = 0.15). Overall survival tended to be longer in patients without versus with PSMA expression (median overall survival: 4.2 vs. 1.9 years; P = 0.273). FGF14 (fibroblast growth factor 14) mRNA expression correlated positively (rho = 0.70; P = 1.70 × 10-5 ) and MAD1L1 (Mitotic spindle assembly checkpoint protein MAD1) correlated negatively with PSMA expression (rho = -0.753; P = 1.58 × 10-6 ). Of the 190 patients who met the eligibility criteria, 31 patients with 39 HCC lesions completed PET; 64% (n = 25) lesions had pronounced 68 Ga-PSMA-11 standardized uptake value: SUVmax (median [range] 9.2 [4.9-28.4]), SUVmean 4.7 (2.4-12.7), and tumor-to-liver background ratio 2 (1.1-11). Conclusion: Ex vivo expression of PSMA in neovasculature of HCC translates to marked tumor avidity on 68 Ga-PSMA-11 PET, which suggests that PSMA has the potential as a theranostic target in patients with HCC.
Collapse
Affiliation(s)
| | - Garima Suman
- Department of RadiologyMayo ClinicRochesterMNUSA
| | | | - Zong‐Ming E. Chen
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMNUSA
| | | | | | | | | | | | | | | | - Lewis R. Roberts
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMNUSA
| | - Kymberly D. Watt
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMNUSA
| | - Mark J. Truty
- Division of Hepatobiliary and Pancreas SurgeryMayo ClinicRochesterMNUSA
| | - Sean P. Cleary
- Division of Hepatobiliary and Pancreas SurgeryMayo ClinicRochesterMNUSA
| | - Rory L. Smoot
- Division of Hepatobiliary and Pancreas SurgeryMayo ClinicRochesterMNUSA
| | | | | | - Amit Mahipal
- Division of Medical OncologyMayo ClinicRochesterMNUSA
| | - Jun Yin
- Division of Biostatistics and InformaticsMayo ClinicRochesterMNUSA
| | - Tyler Zemla
- Division of Biostatistics and InformaticsMayo ClinicRochesterMNUSA
| | - Chen Wang
- Division of Computational BiologyMayo ClinicRochesterMNUSA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Sergeeva O, Zhang Y, Julian W, Sasikumar A, Awadallah A, Kenyon J, Shi W, Sergeev M, Huang S, Sexton S, Iyer R, Xin W, Avril N, Chan ER, Lee Z. Imaging of Tumor-Associated Vascular Prostate-Specific Membrane Antigen in Woodchuck Model of Hepatocellular Carcinoma. GASTRO HEP ADVANCES 2022; 1:631-639. [PMID: 35844243 PMCID: PMC9280909 DOI: 10.1016/j.gastha.2022.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
BACKGROUND AND AIMS Radiolabeled short peptide ligands targeting prostate-specific membrane antigen (PSMA) were developed initially for imaging and treatment of prostate cancers. While many nonprostate solid tumors including hepatocellular carcinoma (HCC) express little PSMA, their neovasculature expresses a high level of PSMA, which is avid for Gallium-68-labeled PSMA-targeting radio-ligand (68Ga-PSMA-11) for positron emission tomography (PET). However, the lack of a spontaneous animal model of tumor-associated vascular PSMA overexpression has hindered the development and assessment of PSMA-targeting radioligands for imaging and therapy of the nonprostatic cancers. We identified detectable indigenous PSMA expression on tumor neovascular endothelia in a naturally occurring woodchuck model of HCC. METHODS Molecular docking was performed with 3 bait PSMA ligands and compared between human and woodchuck PSMA. Initially, PET images were acquired dynamically after intravenously injecting 37 MBq (1.0 mCi) of 68Ga-PSMA-11 into woodchuck models of HCC. Subsequently, 10-minute static PET scans were conducted for other animals 1-hour after injection due to HCC and liver background uptake stabilization at 30-45 minutes after injection. Liver tissue samples were harvested after imaging, fresh-frozen for quantitative reverse transcription polymerase chain reaction and western blot for validation, or fixed for histology for correlation. RESULTS Our preclinical studies confirmed the initial clinical findings of 68Ga-PSMA-11 uptake in HCC. The agents (ligands and antibodies) developed against human PSMA were found to be reactive against the woodchuck PSMA. CONCLUSION This animal model offers a unique opportunity for investigating the biogenesis of tumor-associated vascular PSMA, its functional role(s), and potentials for future treatment strategies targeting tumor vascular PSMA using already developed PSMA-targeting agents.
Collapse
Affiliation(s)
- Olga Sergeeva
- Radiology, Case Western Reserve University, Cleveland, Ohio
| | - Yifan Zhang
- Radiology, Case Western Reserve University, Cleveland, Ohio
| | - Willian Julian
- Radiology, Case Western Reserve University, Cleveland, Ohio
| | - Arun Sasikumar
- Nuclear Medicine, St. Gregorios International Cancer Care Centre, Parumala, Kerala, India
| | - Amad Awadallah
- Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | | | - Wuxian Shi
- Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, Ohio
| | - Maxim Sergeev
- Radiology, University Hospitals Clevel and Medical Center, Cleveland, Ohio
| | - Steve Huang
- Nuclear Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Sandra Sexton
- Medical Oncology, Roswell Park Cancer Institute, Buffalo, New York
| | - Renuka Iyer
- Medical Oncology, Roswell Park Cancer Institute, Buffalo, New York
| | - Wei Xin
- Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Norbert Avril
- Radiology, University Hospitals Clevel and Medical Center, Cleveland, Ohio
| | - Ernest Ricky Chan
- Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio
| | - Zhenghong Lee
- Radiology, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
44
|
Incremental Value of 18F-PSMA-1007 PET/CT in Detection of Metastatic Renal Cell Carcinoma to the Brain. Clin Nucl Med 2022; 47:627-628. [DOI: 10.1097/rlu.0000000000004162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
45
|
Marafi F, Esmail A, Usmani S. Prostate-Specific Membrane Antigen Expression in Metastatic Angiosarcoma Detected on 18F-PMSA PET/CT: A New Potential Prognostic Marker. Clin Nucl Med 2022; 47:e147-e148. [PMID: 34172597 DOI: 10.1097/rlu.0000000000003780] [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: 11/26/2022]
Abstract
ABSTRACT 18F-PSMA is a promising tracer for both staging and detection of biochemical recurrence in prostate cancer. PSMA is also expressed in the endothelium of tumor-associated neovasculature of various solid malignancies possibly due to tumor-associated angiogenic factors and endothelial cell sprouting. We report a case of a 59-year-old man with known case of prostate cancer underwent 18F-PSMA for restaging. 18F-PSMA PET/CT shows multiple lytic bone lesions with high PSMA expression. Histopathological evaluation showed metastatic angiosarcoma. 18F-PSMA expression in the angiosarcoma can be potentially guided to radionuclide legend therapy.
Collapse
Affiliation(s)
- Fahad Marafi
- From the Department of Nuclear Medicine, Jaber Al-Ahmad Molecular Imaging Center
| | - Abdulredha Esmail
- From the Department of Nuclear Medicine, Jaber Al-Ahmad Molecular Imaging Center
| | | |
Collapse
|
46
|
Perera MP, Thomas PB, Risbridger GP, Taylor R, Azad A, Hofman MS, Williams ED, Vela I. Chimeric Antigen Receptor T-Cell Therapy in Metastatic Castrate-Resistant Prostate Cancer. Cancers (Basel) 2022; 14:cancers14030503. [PMID: 35158771 PMCID: PMC8833489 DOI: 10.3390/cancers14030503] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 12/29/2022] Open
Abstract
Simple Summary Prostate cancer is one of the most frequently diagnosed cancers amongst men worldwide. Treatment for metastatic disease is often in the form of androgen deprivation therapy. However, over the course of treatment affected men may become castrate-resistant. Options for men with metastatic castrate-resistant cancer are limited. This review focuses on the role of chimeric antigen receptor T-cell therapy (CAR-T) in men with metastatic castrate-resistant prostate cancer. This review is a contemporary appraisal of preclinical and clinical studies conducted in this emerging form of immunotherapy. A thorough evaluation of the role of CAR-T therapy in prostate cancer is provided, as well as the obstacles we must overcome to clinically translate this therapy for men affected with this rapidly fatal disease. Abstract Prostate cancer is the most commonly diagnosed solid-organ cancer amongst males worldwide. Metastatic castrate-resistant prostate cancer (mCRPC) is a rapidly fatal end-sequelae of prostate cancer. Therapeutic options for men with mCRPC are limited and are not curative in nature. The recent development of chimeric antigen receptor T-cell (CAR-T) therapy has revolutionised the treatment of treatment-resistant haematological malignancies, and several studies are underway investigating the utility of this technology in the treatment of solid tumours. In this review, we evaluate the current treatment options for men with mCRPC as well as the current landscape of preclinical and clinical trials of CAR-T cell therapy against prostate cancer. We also appraise the various prostate cancer-specific tumour-associated antigens that may be targeted by CAR-T cell technology. Finally, we examine the potential translational barriers of CAR-T cell therapy in solid tumours. Despite preclinical success, preliminary clinical trials in men with prostate cancer have had limited efficacy. Therefore, further clinically translatable preclinical models are required to enhance the understanding of the role of this investigational therapeutic in men with mCRPC. In the era of precision medicine, tailored immunotherapy administered to men in a tumour-agnostic approach provides hope to a group of men who otherwise have few treatment options available.
Collapse
Affiliation(s)
- Mahasha P.J. Perera
- School of Biomedical Sciences at Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD 4102, Australia; (P.B.T.); (E.D.W.)
- Queensland Bladder Cancer Initiative (QBCI), Woolloongabba, QLD 4102, Australia
- Department of Urology, Princess Alexandra Hospital, Brisbane, QLD 4102, Australia
- Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD 4102, Australia
- Correspondence: (M.P.P.); (I.V.)
| | - Patrick B. Thomas
- School of Biomedical Sciences at Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD 4102, Australia; (P.B.T.); (E.D.W.)
- Queensland Bladder Cancer Initiative (QBCI), Woolloongabba, QLD 4102, Australia
- Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD 4102, Australia
| | - Gail P. Risbridger
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3168, Australia; (G.P.R.); (R.T.)
| | - Renea Taylor
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3168, Australia; (G.P.R.); (R.T.)
| | - Arun Azad
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (A.A.); (M.S.H.)
| | - Michael S. Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (A.A.); (M.S.H.)
| | - Elizabeth D. Williams
- School of Biomedical Sciences at Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD 4102, Australia; (P.B.T.); (E.D.W.)
- Queensland Bladder Cancer Initiative (QBCI), Woolloongabba, QLD 4102, Australia
- Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD 4102, Australia
| | - Ian Vela
- School of Biomedical Sciences at Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD 4102, Australia; (P.B.T.); (E.D.W.)
- Queensland Bladder Cancer Initiative (QBCI), Woolloongabba, QLD 4102, Australia
- Department of Urology, Princess Alexandra Hospital, Brisbane, QLD 4102, Australia
- Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD 4102, Australia
- Correspondence: (M.P.P.); (I.V.)
| |
Collapse
|
47
|
Unger C, Bronsert P, Michalski K, Bicker A, Juhasz-Böss I. Expression of Prostate Specific Membrane Antigen (PSMA) in Breast Cancer. Geburtshilfe Frauenheilkd 2022; 82:50-58. [PMID: 35027860 PMCID: PMC8747897 DOI: 10.1055/a-1638-9429] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 09/06/2021] [Indexed: 10/28/2022] Open
Abstract
Background Prostate specific membrane antigen (PSMA) is a promising protein for breast cancer patients. It has not only been detected in prostate cancer but is also expressed by tumor cells and the endothelial cells of tumor vessels in breast cancer patients. PSMA plays a role in tumor progression and tumor angiogenesis. For this reason, a number of diagnostic and therapeutic methods to target PSMA have been developed. Method This paper provides a general structured overview of PSMA and its oncogenic potential, with a special focus on its role in breast cancer. This narrative review is based on a selective literature search carried out in PubMed and the library of Freiburg University Clinical Center. The following key words were used for the search: "PSMA", "PSMA and breast cancer", "PSMA PET/CT", "PSMA tumor progression". Relevant articles were explicitly read through, processed, and summarized. Conclusion PSMA could be a new diagnostic and therapeutic alternative, particularly for triple-negative breast cancer. It appears to be a potential predictive and prognostic marker.
Collapse
Affiliation(s)
- Clara Unger
- Klinik für Frauenheilkunde, Universitätsklinikum Freiburg, Freiburg, Germany
| | - Peter Bronsert
- Institut für Klinische Pathologie, Universitätsklinikum Freiburg, Freiburg, Germany
| | - Kerstin Michalski
- Klinik für Nuklearmedizin, Universitätsklinikum Freiburg, Freiburg, Germany
| | - Anna Bicker
- Klinik für Gynäkologie und Geburtshilfe in den St. Vincentius Kliniken, Karslruhe, Germany
| | - Ingolf Juhasz-Böss
- Klinik für Frauenheilkunde, Universitätsklinikum Freiburg, Freiburg, Germany
| |
Collapse
|
48
|
Targeting the IGF-1R in prostate and colorectal cancer: reasons behind trial failure and future directions. Ther Deliv 2022; 13:167-186. [PMID: 35029130 DOI: 10.4155/tde-2021-0060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
IGF-1Rs enact a significant part in cancer growth and its progress. IGF-1R inhibitors were encouraged in the early trials, but the patients did not benefit due to the unavailability of predictive biomarkers and IGF-1R system complexity. However, the linkage between IGF-1R and cancer was reported three decades ago. This review will shed light on the IGF-1R system, targeting IGF-1R through monoclonal antibodies, reasons behind IGF-1R trial failure and future directions. This study presented that targeting IGF-1R through monoclonal antibodies is still effective in cancer treatment, and there is a need to look for future directions. Cancer patients may benefit from using mAbs that target existing and new cancer targets, evidenced by promising results. It is also essential that the academician, trial experts and pharmaceutical companies play their role in finding a treatment for this deadly disease.
Collapse
|
49
|
Abd El Khalek SM, Hafez F. Prostate-specific membrane antigen expression in clear-cell renal cell carcinoma: An angiogenic marker with clinicopathologic significance. EGYPTIAN JOURNAL OF PATHOLOGY 2022; 42:11. [DOI: 10.4103/egjp.egjp_54_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
50
|
Watanabe R, Maekawa M, Kiyoi T, Kurata M, Miura N, Kikugawa T, Higashiyama S, Saika T. PSMA-positive membranes secreted from prostate cancer cells have potency to transform vascular endothelial cells into an angiogenic state. Prostate 2021; 81:1390-1401. [PMID: 34516672 PMCID: PMC9292811 DOI: 10.1002/pros.24237] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/19/2021] [Accepted: 08/30/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND Prostate-specific membrane antigen (PSMA) is highly expressed in poorly differentiated, metastatic, and castration-resistant prostate cancers. Recently, 68Ga-PSMA positron emission tomography/computed tomography has been successfully developed as an effective diagnostic tool for prostate cancer. However, the pathophysiological functions of PSMA in prostate tumors remain unclear. METHODS We examined the protein expression of PSMA in tumor endothelial cells in human prostate tumors by immunohistochemistry. Prostate cancer tissues were resected by robotic surgery in 2019 at Ehime University from patients with prostate cancer. In vitro, we prepared conditioned medium (CM) derived from a PSMA-positive human prostate cancer cell line, LNCaP, cultured on collagen I gels. We then examined PSMA expression in human umbilical vascular endothelial cells (HUVECs) cultured with the CM. We assessed angiogenic activities by treatment of HUVECs with LNCaP-derived CM using a tube formation assay that mimics angiogenesis. RESULTS Immunohistochemistry of PSMA and CD31, a marker of endothelial cells, and PSMA-expressing tumor endothelial cells were observed in 4 of 33 prostate cancer patients (12.1%). We also found that the 10,000g pellet fraction of the LNCaP-derived CM containing PSMA-positive membranes, such as microvesicles transformed HUVECs "PSMA-negative" into "PSMA-positive." Furthermore, treatment of HUVECs with the 10,000g pellet fraction of the LNCaP-derived CM significantly promoted tube formation, mimicking angiogenesis in a PSMA-dependent manner. CONCLUSIONS Our findings revealed the existence of PSMA-positive tumor endothelial cells in human prostate tumors, which enhances tumor angiogenesis in prostate cancer tissues.
Collapse
Affiliation(s)
- Ryuta Watanabe
- Department of UrologyEhime University Graduate School of MedicineMatsuyamaEhimeJapan
- Department of Biochemistry and Molecular GeneticsEhime University Graduate School of MedicineMatsuyamaEhimeJapan
| | - Masashi Maekawa
- Department of Biochemistry and Molecular GeneticsEhime University Graduate School of MedicineMatsuyamaEhimeJapan
- Division of Cell Growth and Tumor Regulation, Proteo‐Science CenterEhime UniversityMatsuyamaEhimeJapan
| | - Takeshi Kiyoi
- Division of Analytical Bio‐medicine, Advanced Research Support CenterEhime UniversityEhimeJapan
| | - Mie Kurata
- Department of PathologyEhime University Graduate School of Medicine and Proteo‐Science CenterEhimeJapan
| | - Noriyoshi Miura
- Department of UrologyEhime University Graduate School of MedicineMatsuyamaEhimeJapan
| | - Tadahiko Kikugawa
- Department of UrologyEhime University Graduate School of MedicineMatsuyamaEhimeJapan
| | - Shigeki Higashiyama
- Department of Biochemistry and Molecular GeneticsEhime University Graduate School of MedicineMatsuyamaEhimeJapan
- Division of Cell Growth and Tumor Regulation, Proteo‐Science CenterEhime UniversityMatsuyamaEhimeJapan
- Department of Molecular and Cellular BiologyOsaka International Cancer InstituteOsakaJapan
| | - Takashi Saika
- Department of UrologyEhime University Graduate School of MedicineMatsuyamaEhimeJapan
| |
Collapse
|