1
|
Van den Broeck B, Debacker JM, Bauters W, Creytens D, Ferdinande L, Huvenne W, Lapauw B, Schelfhout V, Van Laeken N, Verroken C. Potential application of [ 18F]AlF-PSMA-11 PET/CT in radioiodine refractory thyroid carcinoma. EJNMMI Res 2024; 14:82. [PMID: 39264376 PMCID: PMC11393256 DOI: 10.1186/s13550-024-01148-9] [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: 04/19/2024] [Accepted: 08/29/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Patients diagnosed with radioiodine refractory (RAI-R) thyroid carcinoma (TC) have a significantly worse prognosis than patients with radiosensitive TC. These refractory malignancies are often dedifferentiated, hindering the effectiveness of iodine-based imaging. Additionally, the role of metabolic imaging using [18F]FDG PET/CT is also limited in these cases, making adequate staging of RAI-R TC challenging. Recent case series have shown promising results regarding the role of the prostate-specific membrane antigen (PSMA) in TC. In this study we explored the value of [18F]AlF-PSMA-11 PET/CT in RAI-R TC. METHODS In this phase II study, lesions detected on [18F]AlF-PSMA-11 PET were compared to findings from [18F]FDG PET/CT. Additionally, the serologic soluble prostate-specific membrane antigen (sPSMA) was measured using ELISA. PSMA-expression on tumor tissue in any available resection specimens was analysed with an immunostainer. RESULTS Eight patients were included, with a total of 39 identified lesions based on PET imaging. [18F]AlF-PSMA-11 PET identified 30 of 39 lesions, and [18F]FDG PET identified 33 lesions, leading to a detection rate of 76.9% and 84.6%, respectively. Interestingly, while nine lesions were solely visualized on [18F]FDG, six were uniquely seen on [18F]AlF-PSMA-11 PET. While sPSMA was immeasurable in all female patients, no correlation was found between sPSMA in male patients and disease-related factors. In five out of eight patients immunohistology showed PSMA expression on the primary tumor. CONCLUSIONS Although not all lesions could be visualized, [18F]PSMA-11 PET identified multiple lesions imperceptible on [18F]FDG PET. These results display the potential additional diagnostic role of PSMA-targeted imaging in patients with RAI-R TC. Trial registration number No. EudraCT 2021-000456-19.
Collapse
Affiliation(s)
- Bliede Van den Broeck
- Department of Medical Imaging, Nuclear Medicine, Ghent University Hospital, C. Heymanslaan 10, 9000, Ghent, Belgium.
| | - Jens M Debacker
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Department of Nuclear Medicine, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
- Department of Head and Skin, Head and Neck Surgery Research Group, Ghent University, Ghent, Belgium
| | - Wouter Bauters
- Department of Medical Imaging, Radiology, Ghent University Hospital, Ghent, Belgium
| | - David Creytens
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Liesbeth Ferdinande
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Wouter Huvenne
- Department of Head and Skin, Head and Neck Surgery Research Group, Ghent University, Ghent, Belgium
- Department of Head and Neck Surgery, Ghent University Hospital, Ghent, Belgium
| | - Bruno Lapauw
- Department of Endocrinology, Ghent University Hospital, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Vanessa Schelfhout
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Nuclear Medicine, AZ Sint-Jan Brugge, Brugge, Belgium
| | - Nick Van Laeken
- Department of Medical Imaging, Nuclear Medicine, Ghent University Hospital, C. Heymanslaan 10, 9000, Ghent, Belgium
| | | |
Collapse
|
2
|
Wang Z, Zhu B, Jiang F, Chen X, Wang G, Ding N, Song S, Xu X, Zhang W. Design, synthesis and evaluation of novel prostate-specific membrane antigen-targeted aryl [ 18F]fluorosulfate PET tracers. Bioorg Med Chem 2024; 106:117753. [PMID: 38749342 DOI: 10.1016/j.bmc.2024.117753] [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: 03/12/2024] [Revised: 05/01/2024] [Accepted: 05/06/2024] [Indexed: 05/23/2024]
Abstract
The expression of prostate-specific membrane antigen (PSMA) in prostate cancer is 100-1000 times higher than that in normal tissues, and it has shown great advantages in the diagnosis and treatment of prostate cancer. The combination of PSMA and PET imaging technology based on the principle of metabolic imaging can achieve high sensitivity and high specificity for diagnosis. Due to its suitable half-life (109 min) and good positron abundance (97%), as well as its cyclotron accelerated generation, 18F has the potential to be commercialize, which has attracted much attention. In this article, we synthesized a series of fluorosulfate PET tracers targeting PSMA. All four analogues have shown high affinity to PSMA (IC50 = 1.85-5.15 nM). After the radioisotope exchange labeling, [18F]L9 and [18F]L10 have PSMA specific cellular uptake (0.65 ± 0.04% AD and 1.19 ± 0.03% AD) and effectively accumulated in 22Rv1 xenograft mice model. This study demonstrates that PSMA-1007-based PSMA-targeted aryl [18F]fluorosulfate novel tracers have the potential for PET imaging in tumor tissues.
Collapse
Affiliation(s)
- Zhaolin Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Bin Zhu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Fan Jiang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Xiangping Chen
- PET Center, Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Guangfa Wang
- PET Center, Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Ning Ding
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China.
| | - Xiaoping Xu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China.
| | - Wei Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China.
| |
Collapse
|
3
|
Feng Y, Chiou CA, Stagner AM, Chang YS, Freitag SK. Distinguishing spheno-orbital metastatic prostate cancer mimicking a meningioma using novel 18F-PSMA PET/CT imaging. Orbit 2024:1-6. [PMID: 38526143 DOI: 10.1080/01676830.2024.2318769] [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: 11/07/2023] [Accepted: 02/07/2024] [Indexed: 03/26/2024]
Abstract
A 78-year-old man presented with acute-onset left temporal pain, eyelid swelling, and double vision. Computed tomography (CT) demonstrated a left sphenoid wing mass with extra-osseous intra-orbital and intracranial extension, thought to be a typical sphenoid wing meningioma by the primary team. The patient was admitted for an urgent craniotomy, which was planned for the following day. However, upon consultation with ophthalmic plastic surgery, concern was raised for an alternative diagnosis given the atypical timeline, inflammatory changes, and uncharacteristic imaging findings of mixed lytic and sclerotic bony changes without hyperostosis on CT and extensive peri-lesional dural thickening and enhancement on magnetic resonance imaging. A serum prostate-specific antigen was elevated to 206 ng/mL. Subsequent positron emission tomography (PET)/CT using 18F-fluorodeoxyglucose radiotracer was negative for metastatic disease. A prostate-specific membrane antigen (PSMA) PET/CT was then obtained and demonstrated extensive metastases. An orbital biopsy revealed poorly differentiated prostatic adenocarcinoma. The significant incongruence between the standard PET/CT and PSMA PET/CT highlights the value of this novel advanced radiographic modality in narrowing the differential diagnosis and determining the extent of disease. Findings of widespread metastasis on the PSMA PET/CT ultimately helped to avoid a large, morbid neurosurgical intervention in this patient, allowing for a minimally invasive orbital biopsy to characterize the tumor for therapeutic targeting.
Collapse
Affiliation(s)
- Yilin Feng
- Department of Ophthalmology, Ophthalmic Plastic Surgery, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - Carolina A Chiou
- Department of Ophthalmology, Ophthalmic Plastic Surgery, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - Anna M Stagner
- David G. Cogan Laboratory of Ophthalmic Pathology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - Yuh-Shin Chang
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Suzanne K Freitag
- Department of Ophthalmology, Ophthalmic Plastic Surgery, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Lee HW, Kim E, Na I, Kim CK, Seo SI, Park H. Novel Multiparametric Magnetic Resonance Imaging-Based Deep Learning and Clinical Parameter Integration for the Prediction of Long-Term Biochemical Recurrence-Free Survival in Prostate Cancer after Radical Prostatectomy. Cancers (Basel) 2023; 15:3416. [PMID: 37444526 DOI: 10.3390/cancers15133416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Radical prostatectomy (RP) is the main treatment of prostate cancer (PCa). Biochemical recurrence (BCR) following RP remains the first sign of aggressive disease; hence, better assessment of potential long-term post-RP BCR-free survival is crucial. Our study aimed to evaluate a combined clinical-deep learning (DL) model using multiparametric magnetic resonance imaging (mpMRI) for predicting long-term post-RP BCR-free survival in PCa. A total of 437 patients with PCa who underwent mpMRI followed by RP between 2008 and 2009 were enrolled; radiomics features were extracted from T2-weighted imaging, apparent diffusion coefficient maps, and contrast-enhanced sequences by manually delineating the index tumors. Deep features from the same set of imaging were extracted using a deep neural network based on pretrained EfficentNet-B0. Here, we present a clinical model (six clinical variables), radiomics model, DL model (DLM-Deep feature), combined clinical-radiomics model (CRM-Multi), and combined clinical-DL model (CDLM-Deep feature) that were built using Cox models regularized with the least absolute shrinkage and selection operator. We compared their prognostic performances using stratified fivefold cross-validation. In a median follow-up of 61 months, 110/437 patients experienced BCR. CDLM-Deep feature achieved the best performance (hazard ratio [HR] = 7.72), followed by DLM-Deep feature (HR = 4.37) or RM-Multi (HR = 2.67). CRM-Multi performed moderately. Our results confirm the superior performance of our mpMRI-derived DL algorithm over conventional radiomics.
Collapse
Affiliation(s)
- Hye Won Lee
- Samsung Medical Center, Department of Urology, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Eunjin Kim
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Inye Na
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Chan Kyo Kim
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Seong Il Seo
- Samsung Medical Center, Department of Urology, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Hyunjin Park
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon 16419, Republic of Korea
| |
Collapse
|
5
|
Li B, Duan L, Shi J, Han Y, Wei W, Cheng X, Cao Y, Kader A, Ding D, Wu X, Gao Y. Diagnostic performance of 99mTc-HYNIC-PSMA SPECT/CT for biochemically recurrent prostate cancer after radical prostatectomy. Front Oncol 2022; 12:1072437. [PMID: 36568205 PMCID: PMC9768541 DOI: 10.3389/fonc.2022.1072437] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
Objectives 99mTc-HYNIC-PSMA is a novel technetium-99m-labeled small-molecule inhibitor of prostate-specific membrane antigen (PSMA) for detection of prostate cancer. The present study investigated the diagnostic yield of 99mTc-HYNIC-PSMA Single photon emission computed tomography (SPECT)/CT in 147 patients with biochemically recurrent prostate cancer after radical prostatectomy. Methods 147 patients with biochemical relapse after radical prostatectomy were finally eligible for this retrospective analysis. The median prostate-specific antigen (PSA) level was 8.26 ng/mL (range, 0.22-187.40 ng/mL). Of the 147 patients, 72 patients received androgen deprivation therapy (ADT) at least 6 months before the 99mTc-HYNIC-PSMA SPECT/CT. All patients underwent planar whole-body scans and subsequent SPECT/CT of the thoracic and abdominal regions after intravenous injection of 705 ± 70 MBq of 99mTc-HYNIC-PSMA. Images were evaluated for the presence and location of PSMA-positive lesions, in which SUVmax were also measured. Detection rates were stratified according to PSA levels, ADT and Gleason scores. The relationships between SUVmax and clinical characteristics were analyzed using univariate and multivariable linear regression models for patients with positive findings. Results Of the 147 patients, 99mTc-HYNIC-PSMA SPECT/CT revealed at least one positive lesion in 118 patients with a high detection rate (80.3%). The detection rates were 48.6% (17/35), 85.1% (40/47), 92.1% (35/38), and 96.3% (26/27) at PSA levels of greater than 0.2 to 2, greater than 2 to 5, greater than 5 to 10, and greater than 10 ng/mL, respectively. PSMA SPECT/CT indicated local recurrence, lymph node metastases, bone metastases, and visceral metastases in 14 (9.5%), 73 (49.7%), 48 (32.7%) and 3 (2.0%) patients. The detection rates of local recurrence and metastasis increased with increasing PSA levels. The detection rate was higher in patients treated with ADT than those without (90.3% vs. 70.7%; P =0.0029). In patients with Gleason scores ≥8, detection rate was slightly higher than those with ≤7 (81.7% vs. 78.5%), but not statistically significant (P = 0.6265). Multivariable linear regression analysis showed a significant correlation of PSA levels and ADT with SUVmax (P=0.0005 and P=0.0397). Conclusions 99mTc-HYNIC-PSMA SPECT/CT offers high detection rates for biochemically recurrent prostate cancer after radical prostatectomy. The detection rate and SUVmax were positively correlated with PSA levels and ADT.
Collapse
Affiliation(s)
- Bo Li
- Henan Key Laboratory for Molecular Nuclear Medicine and Translational Medicine, Department of Nuclear Medicine, Henan Provincial People’s Hospital & Zhengzhou University People’s Hospital, Zhengzhou, China,Department of Medical Imaging, Xinjiang Production and Construction Corps 13 division Red Star Hospital, Hami, China
| | - Lili Duan
- Henan Key Laboratory for Molecular Nuclear Medicine and Translational Medicine, Department of Nuclear Medicine, Henan Provincial People’s Hospital & Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Jingqi Shi
- Henan Key Laboratory for Molecular Nuclear Medicine and Translational Medicine, Department of Nuclear Medicine, Henan Provincial People’s Hospital & Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Yunyun Han
- Henan Key Laboratory for Molecular Nuclear Medicine and Translational Medicine, Department of Nuclear Medicine, Henan Provincial People’s Hospital & Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Wei Wei
- Department of Urology Surgery, Henan Provincial People’s Hospital & Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Xiaoliang Cheng
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong Universityl, Xi’an, China
| | - Yong Cao
- Department of Medical Imaging, Xinjiang Production and Construction Corps 13 division Red Star Hospital, Hami, China
| | - Akeban Kader
- Department of Medical Imaging, Xinjiang Production and Construction Corps 13 division Red Star Hospital, Hami, China,*Correspondence: Yongju Gao, ; Xinyu Wu, ; Degang Ding, ; Akeban Kader,
| | - Degang Ding
- Department of Urology Surgery, Henan Provincial People’s Hospital & Zhengzhou University People’s Hospital, Zhengzhou, China,*Correspondence: Yongju Gao, ; Xinyu Wu, ; Degang Ding, ; Akeban Kader,
| | - Xinyu Wu
- Henan Key Laboratory for Molecular Nuclear Medicine and Translational Medicine, Department of Nuclear Medicine, Henan Provincial People’s Hospital & Zhengzhou University People’s Hospital, Zhengzhou, China,*Correspondence: Yongju Gao, ; Xinyu Wu, ; Degang Ding, ; Akeban Kader,
| | - Yongju Gao
- Henan Key Laboratory for Molecular Nuclear Medicine and Translational Medicine, Department of Nuclear Medicine, Henan Provincial People’s Hospital & Zhengzhou University People’s Hospital, Zhengzhou, China,*Correspondence: Yongju Gao, ; Xinyu Wu, ; Degang Ding, ; Akeban Kader,
| |
Collapse
|
6
|
Qu C, Zhang H, Cao H, Tang L, Mo H, Liu F, Zhang L, Yi Z, Long L, Yan L, Wang Z, Zhang N, Luo P, Zhang J, Liu Z, Ye W, Liu Z, Cheng Q. Tumor buster - where will the CAR-T cell therapy 'missile' go? Mol Cancer 2022; 21:201. [PMID: 36261831 PMCID: PMC9580202 DOI: 10.1186/s12943-022-01669-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/26/2022] [Indexed: 11/10/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell (CAR-T cell) therapy based on gene editing technology represents a significant breakthrough in personalized immunotherapy for human cancer. This strategy uses genetic modification to enable T cells to target tumor-specific antigens, attack specific cancer cells, and bypass tumor cell apoptosis avoidance mechanisms to some extent. This method has been extensively used to treat hematologic diseases, but the therapeutic effect in solid tumors is not ideal. Tumor antigen escape, treatment-related toxicity, and the immunosuppressive tumor microenvironment (TME) limit their use of it. Target selection is the most critical aspect in determining the prognosis of patients receiving this treatment. This review provides a comprehensive summary of all therapeutic targets used in the clinic or shown promising potential. We summarize CAR-T cell therapies’ clinical trials, applications, research frontiers, and limitations in treating different cancers. We also explore coping strategies when encountering sub-optimal tumor-associated antigens (TAA) or TAA loss. Moreover, the importance of CAR-T cell therapy in cancer immunotherapy is emphasized.
Collapse
Affiliation(s)
- Chunrun Qu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,XiangYa School of Medicine, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hui Cao
- Department of Psychiatry, The Second People's Hospital of Hunan Province, The Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China.,The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lanhua Tang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haoyang Mo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,XiangYa School of Medicine, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fangkun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,XiangYa School of Medicine, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lifu Long
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,XiangYa School of Medicine, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Luzhe Yan
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Nan Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,One-third Lab, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Weijie Ye
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
7
|
Suthar RR, Agrawal A, Deodhar K, Purandare N, Shah S, Puranik A, Choudhury S, Rangarajan V. Unusual Presentation of Helicobacter pylori Infection as PSMA-Avid Gastric Mass. Clin Nucl Med 2022; 47:e607-e608. [PMID: 35384885 DOI: 10.1097/rlu.0000000000004215] [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: 11/25/2022]
Abstract
ABSTRACT 68 Ga-PSMA PET/CT is one of the most common imaging uses for prostate cancer imaging. Although initially thought to be specific for prostate cancer, there are many evidences that are coming of its concentration in many other neoplastic and nonneoplastic pathologies. Helicobacter pylori is the most common bacteria causing gastric inflammation and usually presents with gastroduodenal ulcer. Here we present one unusual case of benign gastric mass formation caused by H. pylori infection with PSMA concentration.
Collapse
Affiliation(s)
- Ritesh Ramesh Suthar
- From the Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Zhao Y, Simpson BS, Morka N, Freeman A, Kirkham A, Kelly D, Whitaker HC, Emberton M, Norris JM. Comparison of Multiparametric Magnetic Resonance Imaging with Prostate-Specific Membrane Antigen Positron-Emission Tomography Imaging in Primary Prostate Cancer Diagnosis: A Systematic Review and Meta-Analysis. Cancers (Basel) 2022; 14:cancers14143497. [PMID: 35884558 PMCID: PMC9323375 DOI: 10.3390/cancers14143497] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/04/2022] [Accepted: 07/12/2022] [Indexed: 02/01/2023] Open
Abstract
Multiparametric magnetic-resonance imaging (mpMRI) has proven utility in diagnosing primary prostate cancer. However, the diagnostic potential of prostate-specific membrane antigen positron-emission tomography (PSMA PET) has yet to be established. This study aims to systematically review the current literature comparing the diagnostic performance of mpMRI and PSMA PET imaging to diagnose primary prostate cancer. A systematic literature search was performed up to December 2021. Quality analyses were conducted using the QUADAS-2 tool. The reference standard was whole-mount prostatectomy or prostate biopsy. Statistical analysis involved the pooling of the reported diagnostic performances of each modality, and differences in per-patient and per-lesion analysis were compared using a Fisher’s exact test. Ten articles were included in the meta-analysis. At a per-patient level, the pooled values of sensitivity, specificity, and area under the curve (AUC) for mpMRI and PSMA PET/CT were 0.87 (95% CI: 0.83−0.91) vs. 0.93 (95% CI: 0.90−0.96, p < 0.01); 0.47 (95% CI: 0.23−0.71) vs. 0.54 (95% CI: 0.23−0.84, p > 0.05); and 0.84 vs. 0.91, respectively. At a per-lesion level, the pooled sensitivity, specificity, and AUC value for mpMRI and PSMA PET/CT were lower, at 0.63 (95% CI: 0.52−0.74) vs. 0.79 (95% CI: 0.62−0.92, p < 0.001); 0.88 (95% CI: 0.81−0.95) vs. 0.71 (95% CI: 0.47−0.90, p < 0.05); and 0.83 vs. 0.84, respectively. High heterogeneity was observed between studies. PSMA PET/CT may better confirm the presence of prostate cancer than mpMRI. However, both modalities appear comparable in determining the localisation of the lesions.
Collapse
Affiliation(s)
- Yi Zhao
- School of Medicine, Imperial College London, London SW7 2BX, UK
- Correspondence:
| | | | - Naomi Morka
- UCL Medical School, University College London, London WC1E 6BT, UK;
| | - Alex Freeman
- Department of Pathology, University College London Hospitals NHS Foundation Trust, London NW1 2PG, UK;
| | - Alex Kirkham
- Department of Radiology, University College London Hospitals NHS Foundation Trust, London NW1 2PG, UK;
| | - Daniel Kelly
- School of Healthcare Sciences, Cardiff University, Cardiff CF10 3AT, UK;
| | - Hayley C. Whitaker
- UCL Division of Surgery & Interventional Science, University College London, London WC1E 6BT, UK; (H.C.W.); (M.E.); (J.M.N.)
| | - Mark Emberton
- UCL Division of Surgery & Interventional Science, University College London, London WC1E 6BT, UK; (H.C.W.); (M.E.); (J.M.N.)
- Department of Urology, University College London Hospitals NHS Foundation Trust, London NW1 2PG, UK
| | - Joseph M. Norris
- UCL Division of Surgery & Interventional Science, University College London, London WC1E 6BT, UK; (H.C.W.); (M.E.); (J.M.N.)
- Department of Urology, University College London Hospitals NHS Foundation Trust, London NW1 2PG, UK
| |
Collapse
|
9
|
Zhao Y, Morka N, Simpson BSS, Freeman A, Kirkham A, Kelly D, Whitaker HC, Emberton M, Norris JM. Prostate-specific membrane antigen positron emission tomography compared to multiparametric MRI for prostate cancer diagnosis: a protocol for a systematic review and meta-analysis. BMJ Open 2021; 11:e052277. [PMID: 34893484 PMCID: PMC8666885 DOI: 10.1136/bmjopen-2021-052277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
INTRODUCTION The introduction of multiparametric MRI (mpMRI) has improved almost every aspect of the prostate cancer diagnostic pathway. However, the novel imaging technique, prostate-specific membrane antigen positron emission tomography (PSMA PET) may have demonstrable accuracy in detecting and staging prostate cancer. Here, we describe a protocol for a systematic review and meta-analysis comparing mpMRI to PSMA PET for the diagnosis of suspected prostate cancer. METHODS AND ANALYSIS A systematic search of MEDLINE, EMBASE, PubMed and Cochrane databases will be conducted. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines will be followed for screening, data extraction, statistical analysis and reporting. Included papers will be full-text articles providing original data, written in English articles and comparing the use of PSMA PET with mpMRI in the diagnosis of prostate cancer. All studies published between July 1977 and March 2021 will be eligible for inclusion. Study bias and quality will be assessed using Quadas-2 score. To ensure the quality of the reporting of studies, this protocol is written following the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols 2015 checklist. ETHICS AND DISSEMINATION Ethical approval will not be required for this systematic review. Findings will be disseminated through peer-reviewed publications and presentations at both national and international conferences. PROSPERO REGISTRATION NUMBER CRD42021239296.
Collapse
Affiliation(s)
- Yi Zhao
- Imperial College London, London, UK
| | - Naomi Morka
- University College London Medical School, London, UK
| | | | - Alex Freeman
- Department of Histopathology, University College Hospital London, London, UK
| | - Alex Kirkham
- Department of Radiology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Daniel Kelly
- School of Healthcare Sciences, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Hayley C Whitaker
- UCL Division of Surgery and Interventional Science, University College London, London, UK
| | - Mark Emberton
- Division of Surgery and Interventional Sciences, University College London, London, UK
- Department of Urology, University College London Hospital, London, UK
| | - Joseph M Norris
- UCL Division of Surgery & Interventional Science, University College London, London, UK
| |
Collapse
|
10
|
Roumeguère T, Aoun F, Albisinni S, Mjaess G. Antibodies targeting Prostate-Specific Membrane Antigen positive prostate cancer: from diagnostic imaging to theranostics. Curr Opin Oncol 2021; 33:500-506. [PMID: 34230440 DOI: 10.1097/cco.0000000000000767] [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: 01/09/2023]
Abstract
PURPOSE OF REVIEW Targeting Prostate-Specific Membrane Antigen (PSMA) has paved the way for personalized medicine in prostate cancer (PCa) patients. This review aims to highlight the role of PSMA targeting antibodies in PCa, for diagnostic and therapeutic purposes. RECENT FINDINGS PSMA Positron Emission Tomography/Computed Tomography has been a game changer in the diagnosis of PCa in the recent decade. Two anti-PSMA monoclonal antibodies have been studied in PCa: 7E11-C35 (limited use) and J591. J591 antibody was used for diagnostic purposes coupled with different radionuclides. Most importantly, it was combined to numerous therapeutic radionuclides such as Lutetium-177 (177Lu), Yttrium-90 (90Y), Indium-111 (111In), and Actinium-225 (225Ac). It was also conjugated to drugs forming antibody-drug conjugates (e.g. MLN2704 and PSMA-ADC). These compounds were tested in recent phase I/II clinical trials. SUMMARY PSMA targeting antibodies are very promising for further clinical investigation and continue to be a momentous research area, for both imaging and therapeutic settings. Although some clinical trials resulted in unfavorably safety profiles for some antibodies, they validated PSMA as a crucial immunoconjugate target.
Collapse
Affiliation(s)
- Thierry Roumeguère
- Department of Urology, University Clinics of Brussels, Hôpital Erasme
- Department of Urology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Fouad Aoun
- Department of Urology, University Clinics of Brussels, Hôpital Erasme
- Department of Urology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Simone Albisinni
- Department of Urology, University Clinics of Brussels, Hôpital Erasme
- Department of Urology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Georges Mjaess
- Department of Urology, University Clinics of Brussels, Hôpital Erasme
| |
Collapse
|
11
|
Papp L, Spielvogel CP, Grubmüller B, Grahovac M, Krajnc D, Ecsedi B, Sareshgi RAM, Mohamad D, Hamboeck M, Rausch I, Mitterhauser M, Wadsak W, Haug AR, Kenner L, Mazal P, Susani M, Hartenbach S, Baltzer P, Helbich TH, Kramer G, Shariat SF, Beyer T, Hartenbach M, Hacker M. Supervised machine learning enables non-invasive lesion characterization in primary prostate cancer with [ 68Ga]Ga-PSMA-11 PET/MRI. Eur J Nucl Med Mol Imaging 2021; 48:1795-1805. [PMID: 33341915 PMCID: PMC8113201 DOI: 10.1007/s00259-020-05140-y] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/29/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE Risk classification of primary prostate cancer in clinical routine is mainly based on prostate-specific antigen (PSA) levels, Gleason scores from biopsy samples, and tumor-nodes-metastasis (TNM) staging. This study aimed to investigate the diagnostic performance of positron emission tomography/magnetic resonance imaging (PET/MRI) in vivo models for predicting low-vs-high lesion risk (LH) as well as biochemical recurrence (BCR) and overall patient risk (OPR) with machine learning. METHODS Fifty-two patients who underwent multi-parametric dual-tracer [18F]FMC and [68Ga]Ga-PSMA-11 PET/MRI as well as radical prostatectomy between 2014 and 2015 were included as part of a single-center pilot to a randomized prospective trial (NCT02659527). Radiomics in combination with ensemble machine learning was applied including the [68Ga]Ga-PSMA-11 PET, the apparent diffusion coefficient, and the transverse relaxation time-weighted MRI scans of each patient to establish a low-vs-high risk lesion prediction model (MLH). Furthermore, MBCR and MOPR predictive model schemes were built by combining MLH, PSA, and clinical stage values of patients. Performance evaluation of the established models was performed with 1000-fold Monte Carlo (MC) cross-validation. Results were additionally compared to conventional [68Ga]Ga-PSMA-11 standardized uptake value (SUV) analyses. RESULTS The area under the receiver operator characteristic curve (AUC) of the MLH model (0.86) was higher than the AUC of the [68Ga]Ga-PSMA-11 SUVmax analysis (0.80). MC cross-validation revealed 89% and 91% accuracies with 0.90 and 0.94 AUCs for the MBCR and MOPR models respectively, while standard routine analysis based on PSA, biopsy Gleason score, and TNM staging resulted in 69% and 70% accuracies to predict BCR and OPR respectively. CONCLUSION Our results demonstrate the potential to enhance risk classification in primary prostate cancer patients built on PET/MRI radiomics and machine learning without biopsy sampling.
Collapse
Affiliation(s)
- L Papp
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - C P Spielvogel
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Christian Doppler Laboratory for Applied Metabolomics, Vienna, Austria
| | - B Grubmüller
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - M Grahovac
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - D Krajnc
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - B Ecsedi
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - R A M Sareshgi
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - D Mohamad
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - M Hamboeck
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - I Rausch
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - M Mitterhauser
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | - W Wadsak
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - A R Haug
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Christian Doppler Laboratory for Applied Metabolomics, Vienna, Austria
| | - L Kenner
- Christian Doppler Laboratory for Applied Metabolomics, Vienna, Austria
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - P Mazal
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - M Susani
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | | | - P Baltzer
- Department of Biomedical Imaging and Image-guided Therapy, Division of Common General and Pediatric Radiology, Medical University of Vienna, Vienna, Austria
| | - T H Helbich
- Department of Biomedical Imaging and Image-guided Therapy, Division of Common General and Pediatric Radiology, Medical University of Vienna, Vienna, Austria
| | - G Kramer
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - S F Shariat
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - T Beyer
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - M Hartenbach
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - M Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
12
|
Satapathy S, Singh H, Kumar R, Mittal BR. Diagnostic Accuracy of 68Ga-PSMA PET/CT for Initial Detection in Patients With Suspected Prostate Cancer: A Systematic Review and Meta-Analysis. AJR Am J Roentgenol 2021; 216:599-607. [PMID: 32755196 DOI: 10.2214/ajr.20.23912] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND. Early diagnosis is important in the overall management of prostate cancer (PCa). Gallium-68-labeled prostate-specific membrane antigen (PSMA) PET/CT has an established role in the detection of recurrent disease and staging of patients with intermediate- to high-risk PCa. However, only a small number of studies have evaluated its role in the initial diagnosis of PCa. OBJECTIVE. This systematic review was conducted to evaluate the diagnostic performance of 68Ga-PSMA PET/CT in the initial detection of PCa in patients with clinical or biochemical findings suspicious for PCa. EVIDENCE ACQUISITION. This systematic review followed PRISMA guidelines. Searches in PubMed, Scopus, and Embase were conducted using relevant keywords, and articles published through April 30, 2020, were included. Using histopathology results as the reference standard, the numbers of true- and false-positives and true- and false-negatives were extracted. Pooled estimates of diagnostic test accuracy-including sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, and summary ROC (SROC) curve-were generated using bivariate random-effects meta-analysis. EVIDENCE SYNTHESIS. Seven studies comprising 389 patients were included in the systematic review and meta-analysis. The pooled sensitivity, specificity, positive likelihood ratio, and negative likelihood ratio for the initial diagnosis of PCa using 68Ga-PSMA PET/CT were 0.97 (95% CI, 0.90-0.99), 0.66 (95% CI, 0.52-0.78), 2.86 (95% CI, 1.95-4.20), and 0.05 (95% CI, 0.01-0.15), respectively. The test had high accuracy; the area under the SROC curve was 0.91 (95% CI, 0.88-0.93). CONCLUSION. Gallium-68-labeled PSMA PET/CT had excellent sensitivity and negative likelihood ratio in the initial diagnosis of PCa in patients with clinical or biochemical findings suspicious for PCa. CLINICAL IMPACT. Gallium-68-labeled PSMA PET/CT had high diagnostic accuracy for the initial detection of PCa in patients with clinical or biochemical findings suspicious for PCa and has potential utility as a rule-out test for these patients.
Collapse
Affiliation(s)
- Swayamjeet Satapathy
- All authors: Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Sector 12, Chandigarh 160012, India
| | - Harmandeep Singh
- All authors: Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Sector 12, Chandigarh 160012, India
| | - Rajender Kumar
- All authors: Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Sector 12, Chandigarh 160012, India
| | - Bhagwant Rai Mittal
- All authors: Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Sector 12, Chandigarh 160012, India
| |
Collapse
|
13
|
Zhang LL, Li WC, Xu Z, Jiang N, Zang SM, Xu LW, Huang WB, Wang F, Sun HB. 68Ga-PSMA PET/CT targeted biopsy for the diagnosis of clinically significant prostate cancer compared with transrectal ultrasound guided biopsy: a prospective randomized single-centre study. Eur J Nucl Med Mol Imaging 2021; 48:483-492. [PMID: 32734457 PMCID: PMC7835307 DOI: 10.1007/s00259-020-04863-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 05/07/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE 68Ga-prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT) is valuable for detecting primary and recurrent prostatic lesions. This study aimed to evaluate the efficacy of 68Ga-PSMA-11 PET/CT as a triage tool for prostate biopsy (PSMA-TB) and compare with transrectal ultrasound-guided biopsy (TRUS-GB) for the diagnosis of clinically significant prostate cancer (csPCa). METHODS This single-centre study randomly allocated 120 patients with elevated serum prostate-specific antigen (PSA) levels (> 4 ng/ml) to PSMA-PET or TRUS group. Patients with PSMA-avid lesions (SUVmax ≥ 8.0) underwent PSMA-TB via a single-puncture percutaneous transgluteal approach (n = 25), whilst patients with negative PSMA-PET underwent systematic TRUS-GB (n = 35). All patients in the TRUS group underwent TRUS-GB directly (n = 60). RESULTS PCa and csPCa were detected in 26/60 (43.3%) and 24/60 (40.0%) patients in the PSMA-PET group and 19/60 (31.6%) and 15/60 (25.0%) in the TRUS group, respectively. In the PSMA-PET group, the detection rate of PCa and csPCa were significantly higher in PSMA-PET-positive than negative patients (PCa, 23/25 (92.0%) vs 3/35 (8.6%), P < 0.01; csPCa, 22/25 (88.0%) vs 2/35 (5.7%), P < 0.01). PSMA-TB detected significantly more PCa and csPCa than TRUS-GB in the TRUS controls (PCa, 21/25 (84.0%) vs 19/60 (31.6%), P < 0.01; csPCa, 20/25 (80.0%) vs 15/60 (25.0%), P < 0.01). PSMA-PET detected significantly more cases of csPCa amongst patients with PSA 4.0-20.0 ng/ml than TRUS (27.02% vs 8.82%, P < 0.05). No haematuria, urinary retention or pelvic infection was observed after PSMA-TB compare with TRUS-GB. CONCLUSIONS 68Ga-PSMA-11 PET/CT is a feasible imaging technique that may serve as a triage tool for prostate biopsy, and may improve the detection rate of csPCa compared with TRUS-GB, especially in patients with serum PSA 4.0-20.0 ng/ml.
Collapse
Affiliation(s)
- Le-Le Zhang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Wen-Cheng Li
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Zheng Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Nan Jiang
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Shi-Ming Zang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Lu-Wei Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Wen-Bing Huang
- Department of Pathology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Feng Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Hong-Bin Sun
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
14
|
Impact of PSMA PET on management of biochemical recurrent prostate cancer: a systematic review and meta-analysis of prospective studies. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-020-00406-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
15
|
Role of metabolic imaging in diagnosis of primary, metastatic, and recurrent prostate cancer. Curr Opin Oncol 2020; 32:223-231. [PMID: 32195681 DOI: 10.1097/cco.0000000000000625] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
PURPOSE OF REVIEW The present review describes the current role of metabolic imaging techniques such as multiparametric MRI (mpMRI), magnetic resonance spectroscopic imaging (MRSI), hyperpolarized MRSI, and positron emission tomography (PET) in the diagnosis of primary prostate cancer, surveillance of low-grade disease, detection of metastases, and evaluation of biochemical recurrence after therapy. RECENT FINDINGS The natural history of prostate cancer ranges from indolent disease that is optimally monitored by active surveillance, to highly aggressive disease that can be lethal. Current diagnostic methods remain imperfect in noninvasively distinguishing between silent versus aggressive tumors. Hence, there is a high demand for noninvasive imaging techniques that offer insight into biological behavior of prostate cancer cells. Characterization of prostate cancer metabolism is a promising area to provide such insights. SUMMARY Metabolic imaging may allow for greater detection and ultimately characterization of tumor based on aggressiveness and spread. Hence, it has the potential to monitor tumor activity, predict prognostic outcomes, and guide individualized therapies.
Collapse
|
16
|
(Radio)Theranostic Patient Management in Oncology Exemplified by Neuroendocrine Neoplasms, Prostate Cancer, and Breast Cancer. Pharmaceuticals (Basel) 2020; 13:ph13030039. [PMID: 32151049 PMCID: PMC7151671 DOI: 10.3390/ph13030039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 03/01/2020] [Accepted: 03/03/2020] [Indexed: 12/18/2022] Open
Abstract
The role of nuclear medicine in the management of oncological patients has expanded during last two decades. The number of radiopharmaceuticals contributing to the realization of theranostics/radiotheranostics in the context of personalized medicine is increasing. This review is focused on the examples of targeted (radio)pharmaceuticals for the imaging and therapy of neuroendocrine neoplasms (NENs), prostate cancer, and breast cancer. These examples strongly demonstrate the tendency of nuclear medicine development towards personalized medicine.
Collapse
|
17
|
Abrantes AM, Pires AS, Monteiro L, Teixo R, Neves AR, Tavares NT, Marques IA, Botelho MF. Tumour functional imaging by PET. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165717. [PMID: 32035103 DOI: 10.1016/j.bbadis.2020.165717] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/15/2020] [Accepted: 01/30/2020] [Indexed: 12/18/2022]
Abstract
Carcinogenesis is a complex multistep process, characterized by changes at different levels, both genetic and epigenetic, which alter cell metabolism. Positron emission tomography (PET) is a very sensitive image modality that allows to evaluate oncometabolism. PET functionalities are immense, since by labelling a molecule that specifically intervenes in a biochemical regulatory pathway of interest with a positron-emitting radionuclide, we can easily image that pathway. Thus, PET makes possible imaging several metabolic processes and assessing risk prediction, screening, diagnosis, response to therapy, metastization and recurrence. In this paper, we provide an overview of different radiopharmaceuticals developed for PET use in oncology, with a focus on brain tumours, breast cancer, hepatocellular carcinoma, neuroendocrine tumours, bladder cancer and prostate cancer because for these cancer types PET has been shown to be valuable. Most of the described tracers are just used in the research environment, with the aim to assess if these tracers could be able to offer an improvement concerning staging/restaging, characterization and stratification of different types of cancer, as well as therapeutic response assessment. In pursuit of personalized therapy, we briefly discuss the more established metabolic tracers and describe recent work on the development of new radiopharmaceuticals, aware that there will continue to exist diagnostic challenges to face modern cancer medicine.
Collapse
Affiliation(s)
- Ana Margarida Abrantes
- Biophysics Institute, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Clinical Academic Center of Coimbra, 3004-561 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; CNC.IBILI Consortium/Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal.
| | - Ana Salomé Pires
- Biophysics Institute, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Clinical Academic Center of Coimbra, 3004-561 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; CNC.IBILI Consortium/Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal.
| | - Lúcia Monteiro
- Biophysics Institute, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Clinical Academic Center of Coimbra, 3004-561 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ricardo Teixo
- Biophysics Institute, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Clinical Academic Center of Coimbra, 3004-561 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; CNC.IBILI Consortium/Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Rita Neves
- Biophysics Institute, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Clinical Academic Center of Coimbra, 3004-561 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Project Development Office, Department of Mathematics and Computer Science, Eindhoven University of Technology (TU/e), NL-5612 AE Eindhoven, the Netherlands
| | - Nuno Tiago Tavares
- Biophysics Institute, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Clinical Academic Center of Coimbra, 3004-561 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Inês Alexandra Marques
- Biophysics Institute, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Clinical Academic Center of Coimbra, 3004-561 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; CNC.IBILI Consortium/Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria Filomena Botelho
- Biophysics Institute, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Clinical Academic Center of Coimbra, 3004-561 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; CNC.IBILI Consortium/Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal.
| |
Collapse
|
18
|
Hess-Busch Y, Hadaschik B, Hess J. M0CRPC overview of management options. World J Urol 2019; 39:349-356. [PMID: 31691081 DOI: 10.1007/s00345-019-02997-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/20/2019] [Indexed: 12/19/2022] Open
Abstract
Though prostate cancer usually responds to androgen deprivation therapy (ADT) in the beginning, the majority of prostate cancers will develop castration resistance over time. The androgen receptor (AR) pathway is often found to be activated in castration resistant prostate cancer (CRPC). Thus, AR signalling remains a therapeutic target upon the development of CRPC. The term M0CRPC is used when ADT leads to castration resistance and there are no metastases detectable by means of conventional imaging. Until recently, there was no therapeutic standard for this group of patients. With the PROSPER-, SPARTAN- and ARAMIS-studies three large placebo-controlled phase III trials have been published lately that showed a significant benefit in metastasis-free survival in men with M0CRPC and short PSA doubling time (PSADT). The efficacy data are very similar in these studies, meaning that the drugs' safety profiles, final analyses of overall survival and their availability will be more important to help clinicians decide which of these three drugs they use for their patients with M0CRPC.
Collapse
Affiliation(s)
- Y Hess-Busch
- Department of Medical Oncology, Kliniken Essen Mitte, Essen, Germany
| | - B Hadaschik
- Department of Urology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - J Hess
- Department of Urology, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
19
|
Cackowski FC, Taichman RS. Minimal Residual Disease in Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1100:47-53. [PMID: 30411259 DOI: 10.1007/978-3-319-97746-1_3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Detection of minimal residual disease (MRD) in prostate cancer over several decades has greatly informed our understanding of dissemination and recurrence, but has not yet been routinely used in clinical care. Investigators have detected MRD by identification of prostate cancer cells in the bone marrow; termed disseminated tumor cells (DTCs) and blood; termed circulating tumor cells (CTCs). Various techniques including PSA-RT PCR, PSA immunocytochemistry, cytokeratin immunocytochemistry, and immune-magnetic depletion of hematopoietic cells followed by EPCAM based positive selection, have been used. Importantly, detection of DTCs correlates with recurrence. Research into prostate cancer CTCs has intensified recently, but their use in MRD evaluation has been more limited. Investigators are using semi-automated platforms to detect and begin to study prostate cancer CTCs in patients with no evidence of disease. PSA immunocytochemistry also detects CTCs and correlates with recurrence. Emerging technologies have the potential to greatly aid research in this exciting field.
Collapse
Affiliation(s)
- Frank C Cackowski
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
- Department of Medicine, Division of Hematology & Oncology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Russell S Taichman
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.
| |
Collapse
|
20
|
Danielson B, Saad F, So A, Morgan S, Hamilton RJ, Malone S, Park-Wyllie L, Zardan A, Shayegan B. Management algorithms for prostate-specific antigen progression in prostate cancer: Biochemical recurrence after definitive therapy and progression to non-metastatic castrate-resistant prostate cancer. Can Urol Assoc J 2019; 13:420-426. [PMID: 31364976 DOI: 10.5489/cuaj.5600] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Current prostate cancer (PCa) guidelines primarily focus on localized or metastatic PCa. A multidisciplinary genitourinary oncology panel determined that additional guidance focusing on monitoring and management of biochemical recurrence (BCR) following radical therapy and non-metastatic castration-resistant prostate cancer (nmCRPC) was warranted. METHODS The most up-to-date national and international guidelines, consensus statements, and emerging phase 3 trials were identified and used to inform development of algorithms by a multidisciplinary genitourinary oncology panel outlining optimal monitoring and treatment for patients with non-metastatic PCa. RESULTS A total of eight major national and international guidelines/consensus statements published since 2015 and three phase 3 trials were identified. Working group discussions among the multidisciplinary genitourinary oncology panel led to the development of two algorithms: the first addressing management of patients with BCR following radical therapy (post-BCR), and the second addressing management of nmCRPC. The post-BCR algorithm suggests consideration of early salvage treatment in select patients and provides guidance regarding observation vs. intermittent or continuous androgen-deprivation therapy (ADT). The nmCRPC algorithm suggests continued ADT and monitoring for all patients, with consideration of treatment with apalutamide or enzalutamide for patients with high-risk disease (prostate-specific antigen [PSA] doubling time of ≤ 10 months). CONCLUSIONS Two treatment algorithms have been developed to guide the management of non-metastatic PCa and should be considered in the context of local guidelines and practice patterns.
Collapse
Affiliation(s)
- Brita Danielson
- Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| | - Fred Saad
- Centre Hospitalier de l'Université de Montréal, Université de Montréal, Montreal, QC, Canada
| | - Alan So
- Vancouver Prostate Centre, University of British Columbia, BC, Canada
| | - Scott Morgan
- The Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | - Robert J Hamilton
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Shawn Malone
- The Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | | | | | - Bobby Shayegan
- Juravinski Cancer Centre, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
21
|
Abstract
Ga-prostate-specific membrane antigen (PSMA) PET/CT scan for primary staging discovered increased Ga-PSMA uptake in a known anal fistula in a recently diagnosed high-risk prostate cancer patient. The patient had an ongoing history of surgical revisions of complex fistulas in the perianal region, contributing to active inflammation and infection. Recently, reports have demonstrated increased Ga-PSMA uptake in different benign inflammatory conditions. This case demonstrates another case of a benign condition associated with increased Ga-PSMA uptake.
Collapse
|
22
|
|
23
|
Chakravarty R, Siamof CM, Dash A, Cai W. Targeted α-therapy of prostate cancer using radiolabeled PSMA inhibitors: a game changer in nuclear medicine. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2018; 8:247-267. [PMID: 30245917 PMCID: PMC6146164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/13/2018] [Indexed: 06/08/2023]
Abstract
Prostate cancer (PCa) is one of the most common malignancies in men and is a major contributor to cancer related deaths worldwide. Metastatic spread and disease progression under androgen deprivation therapy signify the onset of metastatic castration resistant prostate cancer (mCRPCa)-the lethal form of the disease, which severely deteriorates the quality of life of patients. Over the last decade, tremendous progress has been made toward identifying appropriate molecular targets that could enable efficient in vivo targeting for non-invasive imaging and therapy of mCPRCa. In this context, a promising enzymatic target is prostate specific membrane antigen (PSMA), which is overexpressed on PCa cells, in proportion to the stage and grade of the tumor progression. This is especially relevant for mCRPCa, which has significant overexpression of PSMA. For therapy of mCRPCa, several nuclear medicine clinics all over the world have confirmed that 177Lu-labeled-PSMA enzyme inhibitors (177Lu-PSMA-617 and 177Lu-PSMA I&T) have a favorable dosimetry and convincing therapeutic response. However, ~30% of patients were found to be short or non-responders and dose escalation was severely limited by chronic hematological toxicity. Such limitations could be better overcome by targeted alpha therapy (TAT) which has the potential to bring a paradigm shift in treatment of mCRPCa patients. This concise review presents an overview of the successes and challenges currently faced in TAT of mCRPCa using radiolabeled PSMA inhibitors. The preclinical and clinical data reported to date are quite promising, and it is expected that this therapeutic modality will play a pivotal role in advanced stage PCa management in the foreseeable future.
Collapse
Affiliation(s)
- Rubel Chakravarty
- Radiopharmaceuticals Division, Bhabha Atomic Research CentreMumbai 400085, India
- Homi Bhabha National InstituteAnushaktinagar, Mumbai 400094, India
| | - Cerise M Siamof
- Department of Radiology, University of Wisconsin-MadisonWI 53792-3252, USA
| | - Ashutosh Dash
- Radiopharmaceuticals Division, Bhabha Atomic Research CentreMumbai 400085, India
- Homi Bhabha National InstituteAnushaktinagar, Mumbai 400094, India
| | - Weibo Cai
- Department of Radiology, University of Wisconsin-MadisonWI 53792-3252, USA
- Department of Medical Physics, University of Wisconsin-MadisonWI 53705-2275, USA
- Carbone Cancer Center, University of Wisconsin-MadisonWI 53792-3252, USA
| |
Collapse
|
24
|
Nguyen HG, Conn CS, Kye Y, Xue L, Forester CM, Cowan JE, Hsieh AC, Cunningham JT, Truillet C, Tameire F, Evans MJ, Evans CP, Yang JC, Hann B, Koumenis C, Walter P, Carroll PR, Ruggero D. Development of a stress response therapy targeting aggressive prostate cancer. Sci Transl Med 2018; 10:eaar2036. [PMID: 29720449 PMCID: PMC6045425 DOI: 10.1126/scitranslmed.aar2036] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/24/2018] [Accepted: 04/06/2018] [Indexed: 12/23/2022]
Abstract
Oncogenic lesions up-regulate bioenergetically demanding cellular processes, such as protein synthesis, to drive cancer cell growth and continued proliferation. However, the hijacking of these key processes by oncogenic pathways imposes onerous cell stress that must be mitigated by adaptive responses for cell survival. The mechanism by which these adaptive responses are established, their functional consequences for tumor development, and their implications for therapeutic interventions remain largely unknown. Using murine and humanized models of prostate cancer (PCa), we show that one of the three branches of the unfolded protein response is selectively activated in advanced PCa. This adaptive response activates the phosphorylation of the eukaryotic initiation factor 2-α (P-eIF2α) to reset global protein synthesis to a level that fosters aggressive tumor development and is a marker of poor patient survival upon the acquisition of multiple oncogenic lesions. Using patient-derived xenograft models and an inhibitor of P-eIF2α activity, ISRIB, our data show that targeting this adaptive brake for protein synthesis selectively triggers cytotoxicity against aggressive metastatic PCa, a disease for which presently there is no cure.
Collapse
Affiliation(s)
- Hao G Nguyen
- School of Medicine and Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Crystal S Conn
- School of Medicine and Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, CA 94158, USA.
| | - Yae Kye
- School of Medicine and Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Lingru Xue
- School of Medicine and Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Craig M Forester
- Division of Pediatric Allergy, Immunology and Bone Marrow Transplantation, UCSF, San Francisco, CA 94158, USA
| | - Janet E Cowan
- School of Medicine and Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Andrew C Hsieh
- School of Medicine and Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, CA 94158, USA
| | - John T Cunningham
- School of Medicine and Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Charles Truillet
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, CA 94158, USA
| | - Feven Tameire
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J Evans
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, CA 94158, USA
| | - Christopher P Evans
- Department of Urology, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Joy C Yang
- Department of Urology, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Byron Hann
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA 94158, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peter Walter
- Department of Biochemistry and Biophysics, UCSF, Howard Hughes Medical Institute, San Francisco, CA 94158, USA
| | - Peter R Carroll
- School of Medicine and Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Davide Ruggero
- School of Medicine and Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, CA 94158, USA.
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA 94158, USA
| |
Collapse
|