1
|
Rowe SP, Salavati A, Werner RA, Pienta KJ, Gorin MA, Pomper MG, Solnes LB. 18F-Labeled Radiotracers for Prostate-specific Membrane Antigen. PET Clin 2022; 17:585-593. [DOI: 10.1016/j.cpet.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
2
|
Wu Y, Zhang X, Zhou H, Xu B, Tian J, Sun S, Zhang J. Synthesis, preclinical evaluation, and first-in-human study of Al 18F-PSMA-Q for prostate cancer imaging. Eur J Nucl Med Mol Imaging 2022; 49:2774-2785. [PMID: 35396969 DOI: 10.1007/s00259-022-05775-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/21/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE To investigate the potential of a novel Al18F-labeled PSMA-targeted radiotracer for PCa diagnosis through both preclinical and pilot clinical studies. METHODS Al18F-PSMA-Q was prepared automatically. The binding affinity to PSMA was evaluated in vitro using the 22Rv1 (PSMA +) and PC-3 (PSMA -) cell lines. Pharmacokinetics evaluation, biodistribution study, Micro-PET imaging of Al18F-PSMA-Q in normal mice and tumor-bearing mice, and a comparison with 18F-DCFPyL were performed. PET/CT imaging was performed on 8 healthy volunteers and 20 newly diagnosed PCa patients at 1 h post-injection (p.i.). The biodistribution in human and preliminary diagnostic efficacy of Al18F-PSMA-Q were evaluated, and the radiation dosimetry was estimated using OLINDA/EXM 2.0 software. RESULT Qualified Al18F-PSMA-Q was efficiently prepared with a non-decay-corrected radiochemical yield (RCY) of 22.0-28.3%, a specific activity (SA) of > 50 GBq/μmol. The hydrophilicity was comparably high with a log P value of - 3.69 ± 0.39. Al18F-PSMA-Q was found to bind to PSMA specifically with a Ki value of 17.05 ± 1.14 nM. The distribution and elimination half-lives of Al18F-PSMA-Q were 3.93 min and 14.22 min, respectively, which were shorter than those of 18F-DCFPyL. Micro-PET imaging of Al18F-PSMA-Q can clearly differentiate 22Rv1 tumors from PC-3 tumors and background with a high SUVmax of 2.17 ± 0.42 and a tumor-to-muscle ratio of 84.37 ± 31.62, which were higher than those of 18F-DCFPyL (1.79 ± 0.39 and 13.25 ± 1.65). The uptake of Al18F-PSMA-Q in 22Rv1 cells and tumors can be substantially blocked by 2-PMPA. High level accumulation of Al18F-PSMA-Q was observed in organs physiologically expressing PSMA. Twenty-six tumor lesions were detected in 20 PCa patients, and the mean SUVmax values of primary tumors, lymph node metastasis, bone metastases, and tumor-muscle ratios were 19.71 ± 16.52, 5.11, 31.30 ± 29.85, and 44.77 ± 22.29, respectively. The mean SUVmax of tumors in patients with PSA > 10 ng/mL was significantly higher than that in patients with PSA ≤ 10 ng/mL (25.97 ± 18.64 vs. 10.33 ± 3.74). Meanwhile, the mean SUVmax of tumors in patients with a Gleason score ≥ 8 was significantly higher than that in patients with a Gleason score < 8 (31.85 ± 22.09 vs. 13.18 ± 11.58). The kidneys received the highest estimated dose of 0.098 ± 0.006 mGy/MBq, and the effective dose was calculated as 0.0128 ± 0.007 mGy/MBq. CONCLUSION The novel qualified PSMA-targeted radiotracer Al18F-PSMA-Q was conveniently prepared with favorable yield and SA. The results of preclinical and pilot clinical studies exhibited a high specific uptake in PCa lesions and an excellent tumor-to-background ratio with a reasonable radiation exposure, which indicated the great potential of Al18F-PSMA-Q for PCa imaging. TRIAL REGISTRATION Chinese Clinical trial registry ChiCTR2100053507, Registered 23 November 2021, retrospectively registered.
Collapse
Affiliation(s)
- Yitian Wu
- Department of Nuclear Medicine, Chinese PLA General Hospital, No. 28 Fu-Xing Rd., Beijing, 100853, China
| | - Xiaojun Zhang
- Department of Nuclear Medicine, Chinese PLA General Hospital, No. 28 Fu-Xing Rd., Beijing, 100853, China
| | - Haoxi Zhou
- Department of Nuclear Medicine, Chinese PLA General Hospital, No. 28 Fu-Xing Rd., Beijing, 100853, China
| | - Baixuan Xu
- Department of Nuclear Medicine, Chinese PLA General Hospital, No. 28 Fu-Xing Rd., Beijing, 100853, China
| | - Jiahe Tian
- Department of Nuclear Medicine, Chinese PLA General Hospital, No. 28 Fu-Xing Rd., Beijing, 100853, China
| | - Shuwei Sun
- Department of Nuclear Medicine, Chinese PLA General Hospital, No. 28 Fu-Xing Rd., Beijing, 100853, China
| | - Jinming Zhang
- Department of Nuclear Medicine, Chinese PLA General Hospital, No. 28 Fu-Xing Rd., Beijing, 100853, China.
| |
Collapse
|
3
|
El Fakiri M, Geis NM, Ayada N, Eder M, Eder AC. PSMA-Targeting Radiopharmaceuticals for Prostate Cancer Therapy: Recent Developments and Future Perspectives. Cancers (Basel) 2021; 13:cancers13163967. [PMID: 34439121 PMCID: PMC8393521 DOI: 10.3390/cancers13163967] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary One of the most frequently diagnosed cancer in men is adenocarcinoma of the prostate. Once the disease is metastatic, only very limited treatment options are available, resulting in a very short median survival time of 13 months; however, this reality is gradually changing due to the discovery of prostate-specific membrane antigen (PSMA), a protein that is present in cancerous prostate tissue. Researchers have developed pharmaceuticals specific for PSMA, ranging from antibodies (mAb) to low-molecular weight molecules coupled to beta minus and alpha-emitting radionuclides for their use in targeted radionuclide therapy (TRT). TRT offers the possibility of selectively removing cancer tissue via the emission of radiation or radioactive particles within the tumour. In this article, the major milestones in PSMA ligand research and the therapeutic developments are summarised, together with a future perspective on the enhancement of current therapeutic approaches. Abstract Prostate cancer (PC) is the second most common cancer among men, with 1.3 million yearly cases worldwide. Among those cancer-afflicted men, 30% will develop metastases and some will progress into metastatic castration-resistant prostate cancer (mCRPC), which is associated with a poor prognosis and median survival time that ranges from nine to 13 months. Nevertheless, the discovery of prostate specific membrane antigen (PSMA), a marker overexpressed in the majority of prostatic cancerous tissue, revolutionised PC care. Ever since, PSMA-targeted radionuclide therapy has gained remarkable international visibility in translational oncology. Furthermore, on first clinical application, it has shown significant influence on therapeutic management and patient care in metastatic and hormone-refractory prostate cancer, a disease that previously had remained immedicable. In this article, we provide a general overview of the main milestones in the development of ligands for PSMA-targeted radionuclide therapy, ranging from the firstly developed monoclonal antibodies to the current state-of-the-art low molecular weight entities conjugated with various radionuclides, as well as potential future efforts related to PSMA-targeted radionuclide therapy.
Collapse
Affiliation(s)
- Mohamed El Fakiri
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany; (M.E.F.); (N.M.G.); (N.A.); (A.-C.E.)
- Division of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Nicolas M. Geis
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany; (M.E.F.); (N.M.G.); (N.A.); (A.-C.E.)
- Division of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Nawal Ayada
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany; (M.E.F.); (N.M.G.); (N.A.); (A.-C.E.)
- Division of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Matthias Eder
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany; (M.E.F.); (N.M.G.); (N.A.); (A.-C.E.)
- Division of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-761-270-74220
| | - Ann-Christin Eder
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany; (M.E.F.); (N.M.G.); (N.A.); (A.-C.E.)
- Division of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
4
|
Zhang X, Wu Y, Zeng Q, Xie T, Yao S, Zhang J, Cui M. Synthesis, Preclinical Evaluation, and First-in-Human PET Study of Quinoline-Containing PSMA Tracers with Decreased Renal Excretion. J Med Chem 2021; 64:4179-4195. [PMID: 33783213 DOI: 10.1021/acs.jmedchem.1c00117] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The prostate-specific membrane antigen (PSMA) is considered to be an excellent theranostic target of prostate cancer (PCa). In this study, three 18F-labeled PSMA tracers with a more lipophilic quinoline functional spacer were designed, synthesized, and evaluated based on the Glu-Ureido-Lys binding motif. The effect of structure-related lipophilic difference on distribution and excretion of these tracers in vitro and in vivo (cells, rodent, primate, and human) was investigated by comparing with [18F]DCFPyL. There is no significant correlation between the renal elimination and the lipophilicity of the tracers in all species. However, the higher the lipophilicity of tracer, the higher the radioactivity accumulation in the liver of primate and human, and the less radioactivity is to excrete to the bladder with urine. The screened tracer [18F]8c, with a Ki value of 4.58 nM, displayed notable low bladder retention and demonstrated good imaging properties in patients with PCa.
Collapse
Affiliation(s)
- Xiaojun Zhang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
- Department of Nuclear Medicine, Chinese PLA General Hospital, Beijing 100853, China
| | - Yitian Wu
- Department of Nuclear Medicine, Chinese PLA General Hospital, Beijing 100853, China
| | - Qi Zeng
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Tianxin Xie
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Shulin Yao
- Department of Nuclear Medicine, Chinese PLA General Hospital, Beijing 100853, China
| | - Jinming Zhang
- Department of Nuclear Medicine, Chinese PLA General Hospital, Beijing 100853, China
| | - Mengchao Cui
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
- Center for Advanced Materials Research, Advanced Institute of Natural Sciences, Beijing Normal University at Zhuhai, Zhuhai 519087, China
| |
Collapse
|
5
|
Patel TK, Adhikari N, Amin SA, Biswas S, Jha T, Ghosh B. Small molecule drug conjugates (SMDCs): an emerging strategy for anticancer drug design and discovery. NEW J CHEM 2021. [DOI: 10.1039/d0nj04134c] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mechanisms of how SMDCs work. Small molecule drugs are conjugated with the targeted ligand using pH sensitive linkers which allow the drug molecule to get released at lower lysosomal pH. It helps to accumulate the chemotherapeutic agents to be localized in the tumor environment upon cleaving of the pH-labile bonds.
Collapse
Affiliation(s)
- Tarun Kumar Patel
- Epigenetic Research Laboratory, Department of Pharmacy
- BITS-Pilani
- Hyderabad
- India
| | - Nilanjan Adhikari
- Natural Science Laboratory
- Division of Medicinal and Pharmaceutical Chemistry
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata 700032
| | - Sk. Abdul Amin
- Natural Science Laboratory
- Division of Medicinal and Pharmaceutical Chemistry
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata 700032
| | - Swati Biswas
- Epigenetic Research Laboratory, Department of Pharmacy
- BITS-Pilani
- Hyderabad
- India
| | - Tarun Jha
- Natural Science Laboratory
- Division of Medicinal and Pharmaceutical Chemistry
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata 700032
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy
- BITS-Pilani
- Hyderabad
- India
| |
Collapse
|
6
|
Burt T, Young G, Lee W, Kusuhara H, Langer O, Rowland M, Sugiyama Y. Phase 0/microdosing approaches: time for mainstream application in drug development? Nat Rev Drug Discov 2020; 19:801-818. [PMID: 32901140 DOI: 10.1038/s41573-020-0080-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
Abstract
Phase 0 approaches - which include microdosing - evaluate subtherapeutic exposures of new drugs in first-in-human studies known as exploratory clinical trials. Recent progress extends phase 0 benefits beyond assessment of pharmacokinetics to include understanding of mechanism of action and pharmacodynamics. Phase 0 approaches have the potential to improve preclinical candidate selection and enable safer, cheaper, quicker and more informed developmental decisions. Here, we discuss phase 0 methods and applications, highlight their advantages over traditional strategies and address concerns related to extrapolation and developmental timelines. Although challenges remain, we propose that phase 0 approaches be at least considered for application in most drug development scenarios.
Collapse
Affiliation(s)
- Tal Burt
- Burt Consultancy LLC. talburtmd.com, New York, NY, USA. .,Phase-0/Microdosing Network. Phase-0Microdosing.org, New York, NY, USA.
| | - Graeme Young
- GlaxoSmithKline Research and Development Ltd, Ware, UK
| | - Wooin Lee
- Seoul National University, Seoul, Republic of Korea
| | | | - Oliver Langer
- Medical University of Vienna, Vienna, Austria.,AIT Austrian Institute of Technology GmbH, Vienna, Austria
| | | | | |
Collapse
|
7
|
A comparative study of peptide-based imaging agents [ 68Ga]Ga-PSMA-11, [ 68Ga]Ga-AMBA, [ 68Ga]Ga-NODAGA-RGD and [ 68Ga]Ga-DOTA-NT-20.3 in preclinical prostate tumour models. Nucl Med Biol 2020; 84-85:88-95. [PMID: 32251995 DOI: 10.1016/j.nucmedbio.2020.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Peptide-based imaging agents targeting prostate-specific membrane antigen (PSMA) have revolutionized the evaluation of biochemical recurrence of prostate cancer (PCa) but lacks sensitivity at very low serum prostate specific antigen (PSA) levels. Once recurrence is suspected, other positron emission tomography (PET) radiotracers could be of interest to discriminate between local and distant relapse. We studied [18F]fluorodeoxyglucose ([18F]FDG) targeting glucose metabolism, [18F]fluorocholine ([18F]FCH) targeting membrane metabolism and peptide-based imaging agents [68Ga]Ga-PSMA-11, [68Ga]Ga-AMBA, [68Ga]Ga-NODAGA-RGD and [68Ga]Ga-DOTA-NT-20.3 targeting PSMA, gastrin releasing peptide receptor (GRPr), αvβ3 integrin and neurotensin type 1 receptor (NTSR1) respectively, in different PCa tumour models. METHODS Mice were xenografted with 22Rv1, an androgen-receptor (AR)-positive, PCa cell line that expresses PSMA and PC3, an AR-negative one that does not express PSMA. PET imaging using the different radiotracers was performed sequentially and the uptake characteristics compared to one other. NTSR1 and PSMA expression levels were analysed in tumours by immunohistochemistry. RESULTS [18F]FDG displayed low but sufficient uptake to visualize PC3 and 22Rv1 derived tumours. We also observed a low efficacy of [18F]FCH PET imaging and a low [68Ga]Ga-NODAGA-RGD tumour uptake in those tumours. As expected, an elevated tumour uptake was obtained for [68Ga]Ga-PSMA-11 in 22Rv1 derived tumour although no uptake was measured in the androgen independent cell line PC3, derived from a bone metastasis of a high-grade PCa. Moreover, in PC3 cell line, we obtained good tumour uptake, high tumour-to-background contrast using [68Ga]Ga-AMBA and [68Ga]Ga-DOTA-NT-20.3. Immunohistochemistry analysis confirmed high NTSR1 expression in PC3 derived tumours and conversely high PSMA expression in 22Rv1 derived tumours. CONCLUSION PET imaging using [68Ga]Ga-AMBA and [68Ga]Ga-DOTA-NT-20.3 demonstrates that GRPr and NTSR1 could represent viable alternative targets for diagnostic or therapeutic applications in PCa with limited PSMA expression levels. More preclinical and clinical studies will follow to explore this potential. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT Peptide-based imaging agents targeting PSMA represent a major progress in the evaluation of biochemical recurrence of PCa but sometimes yield false negative results in some lesions. Continuing efforts have thus been made to evaluate other radiotracers. Our preclinical results suggest that [68Ga]labelled bombesin and neurotensin analogues could serve as alternative PET radiopharmaceuticals for diagnostic or therapy in cases of PSMA-negative PCa.
Collapse
|
8
|
Werner RA, Derlin T, Lapa C, Sheikbahaei S, Higuchi T, Giesel FL, Behr S, Drzezga A, Kimura H, Buck AK, Bengel FM, Pomper MG, Gorin MA, Rowe SP. 18F-Labeled, PSMA-Targeted Radiotracers: Leveraging the Advantages of Radiofluorination for Prostate Cancer Molecular Imaging. Theranostics 2020; 10:1-16. [PMID: 31903102 PMCID: PMC6929634 DOI: 10.7150/thno.37894] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/11/2019] [Indexed: 12/22/2022] Open
Abstract
Prostate-specific membrane antigen (PSMA)-targeted PET imaging for prostate cancer with 68Ga-labeled compounds has rapidly become adopted as part of routine clinical care in many parts of the world. However, recent years have witnessed the start of a shift from 68Ga- to 18F-labeled PSMA-targeted compounds. The latter imaging agents have several key advantages, which may lay the groundwork for an even more widespread adoption into the clinic. First, facilitated delivery from distant suppliers expands the availability of PET radiopharmaceuticals in smaller hospitals operating a PET center but lacking the patient volume to justify an onsite 68Ge/68Ga generator. Thus, such an approach meets the increasing demand for PSMA-targeted PET imaging in areas with lower population density and may even lead to cost-savings compared to in-house production. Moreover, 18F-labeled radiotracers have a higher positron yield and lower positron energy, which in turn decreases image noise, improves contrast resolution, and maximizes the likelihood of detecting subtle lesions. In addition, the longer half-life of 110 min allows for improved delayed imaging protocols and flexibility in study design, which may further increase diagnostic accuracy. Moreover, such compounds can be distributed to sites which are not allowed to produce radiotracers on-site due to regulatory issues or to centers without access to a cyclotron. In light of these advantageous characteristics, 18F-labeled PSMA-targeted PET radiotracers may play an important role in both optimizing this transformative imaging modality and making it widely available. We have aimed to provide a concise overview of emerging 18F-labeled PSMA-targeted radiotracers undergoing active clinical development. Given the wide array of available radiotracers, comparative studies are needed to firmly establish the role of the available 18F-labeled compounds in the field of molecular PCa imaging, preferably in different clinical scenarios.
Collapse
Affiliation(s)
- Rudolf A. Werner
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Germany
| | - Sara Sheikbahaei
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Würzburg, Germany
- Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Frederik L. Giesel
- Department of Nuclear Medicine, University Hospital Heidelberg, INF 400, 69120 Heidelberg, Germany
| | - Spencer Behr
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital Cologne, Germany
| | - Hiroyuki Kimura
- Department of Analytical and Bioinorganic Chemistry, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Andreas K. Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Germany
| | - Frank M. Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Martin G. Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael A. Gorin
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Steven P. Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
9
|
Abstract
As described in more detail in other contributions in this issue of Seminars in Nuclear Medicine, prostate-specific membrane antigen (PSMA) has become one of the most promising molecular targets in nuclear medicine. Due to its overexpression on prostate cancer cells in proportion to the stage and grade of tumour progression, especially in androgen-independent, advanced and metastatic disease, various tracers for the detection and treatment of prostate cancer by means of radioligand imaging, radioligand therapy or radioguided surgery have been developed and transferred to clinical applications. Even though monoclonal antibodies were investigated and introduced as first PSMA-targeted probes, the inherent advantage of fast tumour uptake and rapid excretion of small molecules has shifted the research focus during the last decade to low molecular weight inhibitors with high affinity to PSMA, such as [18F]FDCFPyL, [18F]PSMA-1007, [68Ga]PSMA-HBED, [177Lu]PSMA-617, [177Lu]PSMA-I&T, [99mTc]MIP-1404 or [99mTc]PSMA I&S, to mention only a few. Due to the plethora of such PSMA probes described during the last years, this review aims to give an overview over the specific characteristics of those radiopharmaceuticals that have already found widespread clinical application. In addition, recently introduced concepts such as PSMA-tracers with increased plasma protein binding, are discussed.
Collapse
Affiliation(s)
- Hans-Jürgen Wester
- Chair of Pharmaceutical Radiochemistry, Walther-Meissner-Strasse 3, 85748 Garching, Germany.
| | - Margret Schottelius
- Chair of Pharmaceutical Radiochemistry, Walther-Meissner-Strasse 3, 85748 Garching, Germany
| |
Collapse
|
10
|
Abstract
The current mainstay of treatment in metastatic prostate cancer is based on hormonal manipulations. Standard androgen deprivation therapy and novel androgen axis drugs are commonly well tolerable and can stabilize metastatic hormone-sensitive prostate cancers for years. However, metastatic castration-resistant prostate cancer is still challenging to treat. Except taxanes, prostate cancer presents intrinsic resistance against conventional chemotherapies. The typically elderly patient population excludes more aggressive treatment regimens. First clinical trials evaluating immunotherapy or tyrosine-kinase-inhibitors against prostate cancer failed. In contrast, prostate cancer can be radiosensitive and external beam radiotherapy is effective in localized prostate cancer, thus providing a good rationale for the use of systemic radiopharmaceuticals in the metastatic setting. Beta-particle emitting "bone-seekers" have a long history and are effective as analgesics but do not improve survival because they are limited by red-marrow dose. Alpha emitting 223Radium can be used in a dose that prolongs survival but is restricted to bone-confined patients. Currently radiolabeled high-affinity ligands to the prostate-specific membrane antigen are in clinical evaluation. While radioimmunotherapy approaches were limited by the long circulation time and slow tumor-accumulation of antibodies, low molecular weight PSMA-specific ligands offer an approx. ten-fold improved tumor to red-marrow ratio in comparison to the unspecific bone-seekers. Early clinical studies demonstrate that regarding surrogate markers, such as >50% PSA reduction (60%) and radiologic response (80%), PSMA-therapy exceeds the antitumor activity of all approved or other recently tested compounds; for example, PSA-response was only observed in approx. a total of 10% of patients treated with ipilimumab, sunitinib, cabozantinib, or xofigo, respectively and in approx. 30, 40, 50% of patients treated with abiraterone, cabazitaxel, or enzalutamide. Also progression free and overall survivals of these single-arm studies appear promising when compared to historical controls. Consecutively, the first PSMA-RLT recently advanced into phase-3 (177Lu-PSMA-617; VISION-trial). Future developments aim to avoid off-target radiation by ligand-optimization and to outperform the antitumor activity of beta-emitter PSMA-RLT by labeling with highly focused, high energy transferring alpha-nuclides; however the latter potentially also increasing the risk of side-effects and additional early phase studies are needed to improve treatment protocols. Academically clinical research is developing prognostic tools to improve treatment benefit by selecting the most appropriate patients in advance.
Collapse
Affiliation(s)
- Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany.
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany; Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frederik L Giesel
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
11
|
Rowe SP, Gorin MA, Pomper MG. Imaging of Prostate-Specific Membrane Antigen with Small-Molecule PET Radiotracers: From the Bench to Advanced Clinical Applications. Annu Rev Med 2019; 70:461-477. [DOI: 10.1146/annurev-med-062117-073027] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In recent years, small-molecule inhibitors of prostate-specific membrane antigen (PSMA) labeled with radionuclides that allow for positron emission tomography (PET) imaging have been extensively studied in many clinical contexts in men with prostate cancer (PCa). The high sensitivity and specificity of these agents for identifying sites of PCa has quickly led to their widespread adoption as a de facto clinical standard of care throughout much of the world. PSMA-targeted PET radiotracers have been particularly well-studied in preoperatively staging men with high-risk PCa, evaluating biochemical recurrence following definitive therapy, and guiding metastasis-directed therapy in patients suspected of having oligorecurrent/oligometastatic disease. Furthermore, the expression of PSMA on the tumor neovasculature of many nonprostate malignancies has enabled a burgeoning subfield concentrated on delineating the potential utility of PSMA-targeted PET agents for imaging other cancers. In this review, we highlight the preclinical development of key small molecules that are now being clinically utilized for PCa imaging, discuss the roles of PSMA-targeted agents in guiding patient management, and consider the role these compounds may play in imaging nonprostate cancers.
Collapse
Affiliation(s)
- Steven P. Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA;,
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | - Michael A. Gorin
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA;,
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | - Martin G. Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA;,
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| |
Collapse
|
12
|
Wüstemann T, Haberkorn U, Babich J, Mier W. Targeting prostate cancer: Prostate-specific membrane antigen based diagnosis and therapy. Med Res Rev 2018; 39:40-69. [PMID: 29771460 DOI: 10.1002/med.21508] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/05/2018] [Accepted: 04/16/2018] [Indexed: 12/12/2022]
Abstract
The high incidence rates of prostate cancer (PCa) raise demand for improved therapeutic strategies. Prostate tumors specifically express the prostate-specific membrane antigen (PSMA), a membrane-bound protease. As PSMA is highly overexpressed on malignant prostate tumor cells and as its expression rate correlates with the aggressiveness of the disease, this tumor-associated biomarker provides the possibility to develop new strategies for diagnostics and therapy of PCa. Major advances have been made in PSMA targeting, ranging from immunotherapeutic approaches to therapeutic small molecules. This review elaborates the diversity of PSMA targeting agents while focusing on the radioactively labeled tracers for diagnosis and endoradiotherapy. A variety of radionuclides have been shown to either enable precise diagnosis or efficiently treat the tumor with minimal effects to nontargeted organs. Most small molecules with affinity for PSMA are based on either a phosphonate or a urea-based binding motif. Based on these pharmacophores, major effort has been made to identify modifications to achieve ideal pharmacokinetics while retaining the specific targeting of the PSMA binding pocket. Several tracers have now shown excellent clinical usability in particular for molecular imaging and therapy as proven by the efficiency of theranostic approaches in current studies. The archetypal expression profile of PSMA may be exploited for the treatment with alpha emitters to break radioresistance and thus to bring the power of systemic therapy to higher levels.
Collapse
Affiliation(s)
- Till Wüstemann
- Department for Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Uwe Haberkorn
- Department for Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - John Babich
- Department for Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Walter Mier
- Department for Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
13
|
Schwarzenböck SM, Schmeja P, Kurth J, Souvatzoglou M, Nawroth R, Treiber U, Kundt G, Berndt S, Graham K, Senekowitsch-Schmidtke R, Schwaiger M, Ziegler SI, Dinkelborg L, Wester HJ, Krause BJ. Comparison of [(11)C]Choline ([(11)C]CHO) and [(18)F]Bombesin (BAY 86-4367) as Imaging Probes for Prostate Cancer in a PC-3 Prostate Cancer Xenograft Model. Mol Imaging Biol 2017; 18:393-401. [PMID: 26483088 DOI: 10.1007/s11307-015-0901-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE Carbon-11- and fluorine-18-labeled choline derivatives are commonly used in prostate cancer imaging in the clinical setting for staging and re-staging of prostate cancer. Due to a limited detection rate of established positron emission tomography (PET) tracers, there is a clinical need for innovative tumor-specific PET compounds addressing new imaging targets. The aim of this study was to compare the properties of [(18)F]Bombesin (BAY 86-4367) as an innovative biomarker for prostate cancer imaging targeting the gastrin-releasing peptide receptor and [(11)C]Choline ([(11)C]CHO) in a human prostate tumor mouse xenograft model by small animal PET/X-ray computed tomography (CT). PROCEDURES We carried out a dual-tracer small animal PET/CT study comparing [(18)F]Bombesin and [(11)C]CHO. The androgen-independent human prostate tumor cell line PC-3 was implanted subcutaneously in the flanks of nu/nu NMRI mice (n = 10) (PET/CT measurements of two [(11)C]Choline mice could not be analyzed due to technical reasons). [(18)F]Bombesin and [(11)C]CHO PET/CT imaging was performed about 3-4 weeks after the implantation of PC-3 cells on two separate days. After the intravenous tail vein injection of 14 MBq [(18)F]Bombesin and 37 MBq [(11)C]CHO, respectively, a dynamic study over 60 min was acquired in list mode using an Inveon animal PET/CT scanner (Siemens Medical Solutions). The sequence of [(18)F]Bombesin and [(11)C]CHO was randomized. Image analysis was performed using summed images as well as dynamic data. To calculate static and dynamic tumor-to-muscle (T/M), tumor-to-blood (T/B), liver-to-blood (L/B), and kidney-to-blood (K/B) ratios, 4 × 4 × 4 mm(3) volumes of interest (VOIs) of tumor, muscle (thigh), liver, kidney, and blood derived from transversal slices were used. RESULTS The mean T/M ratio of [(18)F]Bombesin and [(11)C]CHO was 6.54 ± 2.49 and 1.35 ± 0.30, respectively. The mean T/B ratio was 1.83 ± 0.79 for [(18)F]Bombesin and 0.55 ± 0.10 for [(11)C]CHO. The T/M ratio as well as the T/B ratio for [(18)F]Bombesin were significantly higher compared to those for [(11)C]CHO (p < 0.001, respectively). Kidney and liver uptake was statistically significantly lower for [(18)F]Bombesin (K/B 3.41 ± 0.81, L/B 1.99 ± 0.38) compared to [(11)C]CHO [K/B 7.91 ± 1.85 (p < 0.001), L/B 6.27 ± 1.99 (p < 0.001)]. The magnitudes of the time course of T/M and T/B ratios (T/M and T/Bdyn ratios) were statistically significantly different (showing a higher uptake of [(18)F]Bombesin compared to [(11)C]CHO); additionally, also the change of the T/M and T/B ratios over time was significantly different between both tracers in the dynamic analysis (p < 0.001, respectively). Furthermore, there was a statistically significantly different change of the K/B and L/B ratios over time between the two tracers in the dynamic analysis (p = 0.026 and p < 0.001, respectively). CONCLUSIONS [(18)F]Bombesin (BAY 86-4367) visually and semi-quantitatively outperforms [(11)C]CHO in the PC-3 prostate cancer xenograft model. [(18)F]Bombesin tumor uptake was significantly higher compared to [(11)C]CHO. [(18)F]Bombesin showed better imaging properties compared to the clinically utilized [(11)C]CHO due to a higher tumor uptake as well as a lower liver and kidney uptake.
Collapse
Affiliation(s)
- Sarah Marie Schwarzenböck
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany. .,Department of Nuclear Medicine, Rostock University Medical Centre, Gertrudenplatz 1, 18057, Rostock, Germany.
| | - Philipp Schmeja
- Department of Nuclear Medicine, Rostock University Medical Centre, Gertrudenplatz 1, 18057, Rostock, Germany
| | - Jens Kurth
- Department of Nuclear Medicine, Rostock University Medical Centre, Gertrudenplatz 1, 18057, Rostock, Germany
| | - Michael Souvatzoglou
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany.,Department of Nuclear Medicine, Ulm University, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Roman Nawroth
- Department of Urology, Klinikum Rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Uwe Treiber
- Department of Urology, Klinikum Rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Guenther Kundt
- Department of Biostatistics and Informatics, Rostock University Medical Centre, Ernst-Heydemann-Str. 8, 18057, Rostock, Germany
| | - Sandra Berndt
- Global Drug Discovery, Bayer Healthcare, Muellerstr. 178, 13353, Berlin, Germany
| | - Keith Graham
- Global Drug Discovery, Bayer Healthcare, Muellerstr. 178, 13353, Berlin, Germany
| | - Reingard Senekowitsch-Schmidtke
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Sibylle I Ziegler
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | | | - Hans-Jürgen Wester
- Institution of Pharmaceutical Radiochemistry, Technische Universität München, Walther-Meißner-Str. 3, 85748, Garching, Germany
| | - Bernd Joachim Krause
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany.,Department of Nuclear Medicine, Rostock University Medical Centre, Gertrudenplatz 1, 18057, Rostock, Germany
| |
Collapse
|
14
|
Nedrow JR, Latoche JD, Day KE, Modi J, Ganguly T, Zeng D, Kurland BF, Berkman CE, Anderson CJ. Targeting PSMA with a Cu-64 Labeled Phosphoramidate Inhibitor for PET/CT Imaging of Variant PSMA-Expressing Xenografts in Mouse Models of Prostate Cancer. Mol Imaging Biol 2017; 18:402-10. [PMID: 26552656 DOI: 10.1007/s11307-015-0908-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE Prostate-specific membrane antigen (PSMA) is highly up-regulated in prostate tumor cells, providing an ideal target for imaging applications of prostate cancer. CTT-1297 (IC50 = 27 nM) is an irreversible phosphoramidate inhibitor of PSMA that has been conjugated to the CB-TE1K1P chelator for incorporation of Cu-64. The resulting positron emission tomography (PET) agent, [(64)Cu]ABN-1, was evaluated for selective uptake both in vitro and in vivo in PSMA-positive cells of varying expression levels. The focus of this study was to assess the ability of [(64)Cu]ABN-1 to detect and distinguish varying levels of PSMA in a panel of prostate tumor-bearing mouse models. PROCEDURES CTT-1297 was conjugated to the CB-TE1K1P chelator using click chemistry and radiolabeled with Cu-64. Internalization and binding affinity of [(64)Cu]ABN-1 was evaluated in the following cell lines having varying levels of PSMA expression: LNCaP late-passage > LNCaP early passage ≈ C4-2B > CWR22rv1 and PSMA-negative PC-3 cells. PET/X-ray computed tomography imaging was performed in NCr nude mice with subcutaneous tumors of the variant PSMA-expressing cell lines. RESULTS [(64)Cu]ABN-1 demonstrated excellent uptake in PSMA-positive cells in vitro, with ∼80 % internalization at 4 h for each PSMA-positive cell line with uptake (fmol/mg) correlating to PSMA expression levels. The imaging data indicated significant tumor uptake in all models. The biodistribution for late-passage LNCaP (highest PSMA expression) demonstrated the highest specific uptake of [(64)Cu]ABN-1 with tumor-to-muscle and tumor-to-blood ratios of 30 ± 11 and 21 ± 7, respectively, at 24 h post-injection. [(64)Cu]ABN-1 cleared through all tissues except for PSMA-positive kidneys. CONCLUSION [(64)Cu]ABN-1 demonstrated selective uptake in PSMA-positive cells and tumors, which correlated to the level of PSMA expression. The data reported herein suggest that [(64)Cu]ABN-1 will selectively target and image variant PSMA expression and in the future will serve as a non-invasive method to follow the progression of prostate cancer in men.
Collapse
Affiliation(s)
- Jessie R Nedrow
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Joseph D Latoche
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Kathryn E Day
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Jalpa Modi
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Tanushree Ganguly
- Department of Chemistry, Washington State University, Pullman, WA, USA
| | - Dexing Zeng
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Brenda F Kurland
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Carolyn J Anderson
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, 15219, USA.
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
15
|
Rowe SP, Drzezga A, Neumaier B, Dietlein M, Gorin MA, Zalutsky MR, Pomper MG. Prostate-Specific Membrane Antigen-Targeted Radiohalogenated PET and Therapeutic Agents for Prostate Cancer. J Nucl Med 2017; 57:90S-96S. [PMID: 27694179 DOI: 10.2967/jnumed.115.170175] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 08/08/2016] [Indexed: 12/16/2022] Open
Abstract
Radiohalogenated agents are often the first line of pursuit in the development of new radiopharmaceuticals-whether antibodies, peptides, or small molecules-because of their ease of synthesis, lack of substantial steric perturbation of the original affinity agent (in some cases, providing enhanced affinity), and capacity to be transformed into therapeutics (in some cases, with a mere switch of an isotope). They often provide proof of a principle before optimization for pharmacokinetics or generation of radiometallated agents, when the latter are necessary. In particular, 18F has been well integrated into normal clinical work flow in the form of 18F-FDG for oncologic imaging, with reliable daily production and distribution to sites for immediate use, without the need for on-site preparation. Here we discuss radiohalogenated versions of imaging and therapeutic agents targeting the prostate-specific membrane antigen (PSMA); these were among the first such agents to be synthesized and used clinically. PSMA is highly expressed on prostate cancer epithelial cells and is currently being extensively investigated around the world as a target for imaging and therapy of prostate cancer. Additionally, the presence of PSMA on nonprostate tumor neovasculature has opened the possibility of PSMA-targeted molecules as generalizable cancer imaging and therapy agents. We focus on 18F-labeled agents for PET, as they begin to redefine-along with the corresponding 68Ga-labeled agents discussed elsewhere in this supplement to The Journal of Nuclear Medicine-the management of prostate cancer across a variety of clinical contexts.
Collapse
Affiliation(s)
- Steven P Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital of Cologne, Cologne, Germany
| | - Bernd Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, University Hospital of Cologne, Cologne, Germany
| | - Markus Dietlein
- Department of Nuclear Medicine, University Hospital of Cologne, Cologne, Germany
| | - Michael A Gorin
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Michael R Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
16
|
Gourni E, Henriksen G. Metal-Based PSMA Radioligands. Molecules 2017; 22:molecules22040523. [PMID: 28338640 PMCID: PMC6154343 DOI: 10.3390/molecules22040523] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/13/2017] [Accepted: 03/18/2017] [Indexed: 12/20/2022] Open
Abstract
Prostate cancer is one of the most common malignancies for which great progress has been made in identifying appropriate molecular targets that would enable efficient in vivo targeting for imaging and therapy. The type II integral membrane protein, prostate specific membrane antigen (PSMA) is overexpressed on prostate cancer cells in proportion to the stage and grade of the tumor progression, especially in androgen-independent, advanced and metastatic disease, rendering it a promising diagnostic and/or therapeutic target. From the perspective of nuclear medicine, PSMA-based radioligands may significantly impact the management of patients who suffer from prostate cancer. For that purpose, chelating-based PSMA-specific ligands have been labeled with various diagnostic and/or therapeutic radiometals for single-photon-emission tomography (SPECT), positron-emission-tomography (PET), radionuclide targeted therapy as well as intraoperative applications. This review focuses on the development and further applications of metal-based PSMA radioligands.
Collapse
Affiliation(s)
- Eleni Gourni
- Institute of Basic Medical Sciences, University of Oslo, Oslo 0372, Norway.
- Norwegian Medical Cyclotron Centre Ltd., P.O. Box 4950 Nydalen, Oslo 0424, Norway.
| | - Gjermund Henriksen
- Institute of Basic Medical Sciences, University of Oslo, Oslo 0372, Norway.
- Norwegian Medical Cyclotron Centre Ltd., P.O. Box 4950 Nydalen, Oslo 0424, Norway.
- Institute of Physics, University of Oslo, Oslo 0317, Norway.
| |
Collapse
|
17
|
Evans JC, Malhotra M, Cryan JF, O'Driscoll CM. The therapeutic and diagnostic potential of the prostate specific membrane antigen/glutamate carboxypeptidase II (PSMA/GCPII) in cancer and neurological disease. Br J Pharmacol 2016; 173:3041-3079. [PMID: 27526115 PMCID: PMC5056232 DOI: 10.1111/bph.13576] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 07/08/2016] [Accepted: 07/25/2016] [Indexed: 12/11/2022] Open
Abstract
Prostate specific membrane antigen (PSMA) otherwise known as glutamate carboxypeptidase II (GCPII) is a membrane bound protein that is highly expressed in prostate cancer and in the neovasculature of a wide variety of tumours including glioblastomas, breast and bladder cancers. This protein is also involved in a variety of neurological diseases including schizophrenia and ALS. In recent years, there has been a surge in the development of both diagnostics and therapeutics that take advantage of the expression and activity of PSMA/GCPII. These include gene therapy, immunotherapy, chemotherapy and radiotherapy. In this review, we discuss the biological roles that PSMA/GCPII plays, both in normal and diseased tissues, and the current therapies exploiting its activity that are at the preclinical stage. We conclude by giving an expert opinion on the future direction of PSMA/GCPII based therapies and diagnostics and hurdles that need to be overcome to make them effective and viable.
Collapse
Affiliation(s)
- James C Evans
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - Meenakshi Malhotra
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | | |
Collapse
|
18
|
Pillai MRA, Nanabala R, Joy A, Sasikumar A, Russ Knapp FF. Radiolabeled enzyme inhibitors and binding agents targeting PSMA: Effective theranostic tools for imaging and therapy of prostate cancer. Nucl Med Biol 2016; 43:692-720. [PMID: 27589333 DOI: 10.1016/j.nucmedbio.2016.08.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 08/09/2016] [Accepted: 08/09/2016] [Indexed: 12/14/2022]
Abstract
Because of the broad incidence, morbidity and mortality associated with prostate-derived cancer, the development of more effective new technologies continues to be an important goal for the accurate detection and treatment of localized prostate cancer, lymphatic involvement and metastases. Prostate-specific membrane antigen (PSMA; Glycoprotein II) is expressed in high levels on prostate-derived cells and is an important target for visualization and treatment of prostate cancer. Radiolabeled peptide targeting technologies have rapidly evolved over the last decade and have focused on the successful development of radiolabeled small molecules that act as inhibitors to the binding of the N-acetyl-l-aspartyl-l-glutamate (NAAG) substrate to the PSMA molecule. A number of radiolabeled PSMA inhibitors have been described in the literature and labeled with SPECT, PET and therapeutic radionuclides. Clinical studies with these agents have demonstrated the improved potential of PSMA-targeted PET imaging agents to detect metastatic prostate cancer in comparison with conventional imaging technologies. Although many of these agents have been evaluated in humans, by far the most extensive clinical literature has described use of the 68Ga and 177Lu agents. This review describes the design and development of these agents, with a focus on the broad clinical introduction of PSMA targeting motifs labeled with 68Ga for PET-CT imaging and 177Lu for therapy. In particular, because of availability from the long-lived 68Ge (T1/2=270days)/68Ga (T1/2=68min) generator system and increasing availability of PET-CT, the 68Ga-labeled PSMA targeted agent is receiving widespread interest and is one of the fastest growing radiopharmaceuticals for PET-CT imaging.
Collapse
Affiliation(s)
| | - Raviteja Nanabala
- KIMS DDNMRC PET Scans, KIMS Hospital, Trivandrum, Kerala, India, 691601
| | - Ajith Joy
- Molecular Group of Companies, Puthuvype, Ernakulam, Kerala, 682508, India
| | - Arun Sasikumar
- KIMS DDNMRC PET Scans, KIMS Hospital, Trivandrum, Kerala, India, 691601
| | - Furn F Russ Knapp
- Emeritus, Medical Radioisotope Program, Oak Ridge National Laboratory, Oak Ridge, TN, USA, 37830
| |
Collapse
|
19
|
Abstract
The use of positron emission tomography (PET) is an established method for the diagnosis of urological malignancies. Several tracers are currently available to obtain metabolic information or directly detect molecular targets. While (18)F-FDG-PET is recognized in current guidelines for the staging of seminoma, PET is not used in clinical routine in renal malignancies due to the lack of specific tracers. Despite initial promising results in bladder cancer, no relevant additional diagnostic value with PET using (18)F-FDG or choline-based tracers could be obtained in most patients and therefore should be used with caution or only within clinical trials. In prostate cancer, however, after development of new tracers that, for example, target prostate-specific membrane antigen (PSMA), a paradigm shift in imaging can be recognized. Here, (68)Ga-PSMA-PET might be included in the future as part of standard imaging work-up.
Collapse
Affiliation(s)
- T Maurer
- Urologische Klinik und Poliklinik, Technische Universität München, Klinikum rechts der Isar, Ismaninger Straße 22, 81671, München, Deutschland,
| | | | | | | |
Collapse
|
20
|
Machulkin AE, Ivanenkov YA, Aladinskaya AV, Veselov MS, Aladinskiy VA, Beloglazkina EK, Koteliansky VE, Shakhbazyan AG, Sandulenko YB, Majouga AG. Small-molecule PSMA ligands. Current state, SAR and perspectives. J Drug Target 2016; 24:679-93. [DOI: 10.3109/1061186x.2016.1154564] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Alexey E. Machulkin
- Moscow State University, Chemistry Dept, Leninskie Gory, Moscow, Russian Federation
- National University of Science and Technology MISiS, Moscow, Russian Federation
| | - Yan A. Ivanenkov
- Moscow State University, Chemistry Dept, Leninskie Gory, Moscow, Russian Federation
- National University of Science and Technology MISiS, Moscow, Russian Federation
- Moscow Institute of Physics and Technology (State University), Dolgoprudny City, Russian Federation
| | - Anastasia V. Aladinskaya
- Moscow Institute of Physics and Technology (State University), Dolgoprudny City, Russian Federation
| | - Mark S. Veselov
- Moscow Institute of Physics and Technology (State University), Dolgoprudny City, Russian Federation
| | - Vladimir A. Aladinskiy
- Moscow Institute of Physics and Technology (State University), Dolgoprudny City, Russian Federation
| | | | - Victor E. Koteliansky
- Moscow State University, Chemistry Dept, Leninskie Gory, Moscow, Russian Federation
- Skolkovo Institute of Science and Technology “Scoltech”, Novaya, Russian Federation
| | - Artem G. Shakhbazyan
- Moscow Institute of Physics and Technology (State University), Dolgoprudny City, Russian Federation
| | - Yuri B. Sandulenko
- Moscow Institute of Physics and Technology (State University), Dolgoprudny City, Russian Federation
| | - Alexander G. Majouga
- Moscow State University, Chemistry Dept, Leninskie Gory, Moscow, Russian Federation
- National University of Science and Technology MISiS, Moscow, Russian Federation
| |
Collapse
|
21
|
Benešová M, Bauder-Wüst U, Schäfer M, Klika KD, Mier W, Haberkorn U, Kopka K, Eder M. Linker Modification Strategies To Control the Prostate-Specific Membrane Antigen (PSMA)-Targeting and Pharmacokinetic Properties of DOTA-Conjugated PSMA Inhibitors. J Med Chem 2016; 59:1761-75. [DOI: 10.1021/acs.jmedchem.5b01210] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Martina Benešová
- Division
of Radiopharmaceutical Chemistry, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Ulrike Bauder-Wüst
- Division
of Radiopharmaceutical Chemistry, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Martin Schäfer
- Division
of Radiopharmaceutical Chemistry, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Karel D. Klika
- Molecular
Structure Analysis, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Walter Mier
- Department
of Nuclear Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Uwe Haberkorn
- Department
of Nuclear Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Clinical
Cooperation Unit Nuclear Medicine, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Klaus Kopka
- Division
of Radiopharmaceutical Chemistry, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Matthias Eder
- Division
of Radiopharmaceutical Chemistry, German Cancer Research Center, 69120 Heidelberg, Germany
| |
Collapse
|
22
|
Keck B, Hammon M, Uder M, Huber J, Goebell PJ, Kunath F, Wullich B, Richter RH. [Vertebral metastases of urogenital carcinomas: Diagnosis and conservative therapy]. Urologe A 2015; 55:226-31. [PMID: 26450096 DOI: 10.1007/s00120-015-3977-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The high incidence of bone metastases of urologic neoplasms and their morbidity, especially of vertebral metastases, requires exact diagnosis and consequent therapy. Conventional radiography plays an important role in the diagnosis of symptomatic bone lesions. Computed tomography can evaluate the stability of metastatic lesions and is indispensable for therapy planning. MRI and PET-CT have the highest diagnostic accuracy for the detection of bone metastases and MRI can evaluate their intra- and extraosseus components. PET-CT, PET-MRI, or SPECT-CT in combination with specific tracers - due to their high specificity and sensitivity - have the potential to replace conventional methods in the future. Conservative treatment basically consists of analgesic therapy, the administration of calcium and vitamin D3 and bisphosphonates or inhibitors of RANKL (denosumab). Moreover radium-223-dichloride can improve overall survival and the time to the first symptomatic skeletal event in castration-resistant prostate cancer patients with bone metastases.
Collapse
Affiliation(s)
- B Keck
- Urologische Universitätsklinik Erlangen, Universitätsklinikum Erlangen, Krankenhausstraße 12, 91054, Erlangen, Deutschland.
| | - M Hammon
- Radiologisches Institut, Universitätsklinikum Erlangen, Erlangen, Deutschland
| | - M Uder
- Radiologisches Institut, Universitätsklinikum Erlangen, Erlangen, Deutschland
| | - J Huber
- Klinik und Poliklinik für Urologie, Medizinische Fakultät Carl Gustav Carus, TU Dresden, Dresden, Deutschland
| | - P J Goebell
- Urologische Universitätsklinik Erlangen, Universitätsklinikum Erlangen, Krankenhausstraße 12, 91054, Erlangen, Deutschland
| | - F Kunath
- Urologische Universitätsklinik Erlangen, Universitätsklinikum Erlangen, Krankenhausstraße 12, 91054, Erlangen, Deutschland
| | - B Wullich
- Urologische Universitätsklinik Erlangen, Universitätsklinikum Erlangen, Krankenhausstraße 12, 91054, Erlangen, Deutschland
| | - R H Richter
- Orthopädische Universitätsklinik Erlangen, Friedrich-Alexander-Universität Erlangen, Erlangen, Deutschland
| |
Collapse
|
23
|
Zechmann CM. Imaging for Prostate Cancer. CURRENT RADIOLOGY REPORTS 2015. [DOI: 10.1007/s40134-015-0107-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
24
|
Synthesis, 68Ga-radiolabeling, and preliminary in vivo assessment of a depsipeptide-derived compound as a potential PET/CT infection imaging agent. BIOMED RESEARCH INTERNATIONAL 2015; 2015:284354. [PMID: 25699267 PMCID: PMC4324493 DOI: 10.1155/2015/284354] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Revised: 07/28/2014] [Accepted: 08/05/2014] [Indexed: 11/21/2022]
Abstract
Noninvasive imaging is a powerful tool for early diagnosis and monitoring of various disease processes, such as infections. An alarming shortage of infection-selective radiopharmaceuticals exists for overcoming the diagnostic limitations with unspecific tracers such as 67/68Ga-citrate or 18F-FDG. We report here TBIA101, an antimicrobial peptide derivative that was conjugated to DOTA and radiolabeled with 68Ga for a subsequent in vitro assessment and in vivo infection imaging using Escherichia coli-bearing mice by targeting bacterial lipopolysaccharides with PET/CT. Following DOTA-conjugation, the compound was verified for its cytotoxic and bacterial binding behaviour and compound stability, followed by 68Gallium-radiolabeling. µPET/CT using 68Ga-DOTA-TBIA101 was employed to detect muscular E. coli-infection in BALB/c mice, as warranted by the in vitro results. 68Ga-DOTA-TBIA101-PET detected E. coli-infected muscle tissue (SUV = 1.3–2.4) > noninfected thighs (P = 0.322) > forearm muscles (P = 0.092) > background (P = 0.021) in the same animal. Normalization of the infected thigh muscle to reference tissue showed a ratio of 3.0 ± 0.8 and a ratio of 2.3 ± 0.6 compared to the identical healthy tissue. The majority of the activity was cleared by renal excretion. The latter findings warrant further preclinical imaging studies of greater depth, as the DOTA-conjugation did not compromise the TBIA101's capacity as targeting vector.
Collapse
|
25
|
Beheshti M, Kunit T, Haim S, Zakavi R, Schiller C, Stephens A, Dinkelborg L, Langsteger W, Loidl W. BAY 1075553 PET-CT for Staging and Restaging Prostate Cancer Patients: Comparison with [18F] Fluorocholine PET-CT (Phase I Study). Mol Imaging Biol 2014; 17:424-33. [DOI: 10.1007/s11307-014-0800-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
26
|
Nanni C, Zanoni L, Fanti S. Nuclear medicine in urological cancers: what is new? Future Oncol 2014; 10:2061-72. [DOI: 10.2217/fon.14.87] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
ABSTRACT The diffusion of PET/computed tomography has opened up a new role for nuclear imaging in urological oncology. Prostate cancer is evaluated with choline (11C or 18F) PET due to a lack of sensitivity of 18F-fluorodeoxyglucose (FDG). However, many new tracers, such as 18F-fluorocyclobutane-1-carboxylic acid and 68Ga-prostate-specific membrane antigen, are under investigation, offering promising results in the particular setting of radically treated patients with biochemical relapse. The performance of 18F-FDG depends on the histological type; indeed, renal cell cancer may present variable metabolic uptake. In this field, mainly antibodies labeled with positron emitters are under clinical evaluation. Finally, 18F-FDG PET/computed tomography has been proven to show good accuracy in detecting metastatic testicular and bladder cancers, despite not having valid results in detecting local disease. The urological cancer diagnostic process is currently under continuous development.
Collapse
Affiliation(s)
- Cristina Nanni
- OU Nuclear Medicine, Azienda Ospedaliero-Universitaria di Bologna Policlinico S Orsola-Malpighi, Via Massarenti, 9-40138 Bologna, Italy
| | - Lucia Zanoni
- OU Nuclear Medicine, Azienda Ospedaliero-Universitaria di Bologna Policlinico S Orsola-Malpighi, Via Massarenti, 9-40138 Bologna, Italy
| | - Stefano Fanti
- OU Nuclear Medicine, Azienda Ospedaliero-Universitaria di Bologna Policlinico S Orsola-Malpighi, Via Massarenti, 9-40138 Bologna, Italy
| |
Collapse
|
27
|
Wiehr S, Bühler P, Gierschner D, Wolf P, Rolle AM, Kesenheimer C, Pichler BJ, Elsässer-Beile U. Pharmacokinetics and PET imaging properties of two recombinant anti-PSMA antibody fragments in comparison to their parental antibody. Prostate 2014; 74:743-55. [PMID: 24610028 DOI: 10.1002/pros.22794] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 01/31/2014] [Indexed: 01/10/2023]
Abstract
BACKGROUND Radioimmunoimaging with disease-specific tracers can be advantageous compared to that with nonspecific tracers for the imaging of glucose metabolism and cell proliferation. Monoclonal antibodies (mAbs) or their fragments are excellent tools for immuno-positron emission tomography (PET). In this study, PSMA-specific mAb 3/F11 and its recombinant fragments were compared for the imaging of prostate cancer in xenografts. METHODS Recombinant anti-PSMA antibody fragments D7-Fc and D7-CH3 were constructed by genetically fusing the binding domains of mAb 3/F11 (D7) to the human IgG3 CH3 or CH2-CH3 (Fc) domain. The fragments and the mAb 3/F11 were DOTA conjugated, tested in vitro, and radiolabeled with (64) Cu. PSMA-positive C4-2 and PSMA-negative DU 145 prostate cancer xenografts were used for PET-MR imaging and for ex vivo biodistribution. RESULTS The constructs showed strong and specific binding to PSMA-positive C4-2 cells in vitro which did not decrease after DOTA conjugation. Both tested fragments showed stable accumulation in PSMA-positive C4-2 tumors at all measured time points but reduced uptake compared to the full-length antibody. Other organs and PSMA-negative tumors showed a very low tracer uptake only 3 hr after injection, with the exception of the kidneys, which demonstrated high radioactivity uptake due to rapid renal clearance of the mAb fragments. CONCLUSION Stable tumor uptake and fast serum clearance of the tested radiolabeled fragments was observed in this preclinical study compared to the full length mAb. Since the fragments show rapid and specific tumor uptake, the tested fragments might serve as tools for theranostic imaging with suitable isotopes for radioimmunotherapy.
Collapse
Affiliation(s)
- Stefan Wiehr
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University, Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
There is an expanding and exciting repertoire of PET imaging radiotracers for urogenital diseases, particularly in prostate cancer, renal cell cancer, and renal function. Prostate cancer is the most commonly diagnosed cancer in men. With growing therapeutic options for the treatment of metastatic and advanced prostate cancer, improved functional imaging of prostate cancer beyond the limitations of conventional CT and bone scan is becoming increasingly important for both clinical management and drug development. PET radiotracers, apart from ¹⁸F-FDG, for prostate cancer are ¹⁸F-sodium fluoride, ¹¹C-choline, and ¹⁸F-fluorocholine, and (¹¹C-acetate. Other emerging and promising PET radiotracers include a synthetic l-leucine amino acid analogue (anti-¹⁸F-fluorocyclobutane-1-carboxylic acid), dihydrotestosterone analogue (¹⁸F-fluoro-5α-dihydrotestosterone), and prostate-specific membrane antigen-based PET radiotracers (eg, N-[N-[(S)-1,3-dicarboxypropyl]carbamoyl]-4-¹⁸F-fluorobenzyl-l-cysteine, ⁸⁹Zr-DFO-J591, and ⁶⁸Ga [HBED-CC]). Larger prospective and comparison trials of these PET radiotracers are needed to establish the role of PET/CT in prostate cancer. Although renal cell cancer imaging with FDG-PET/CT is available, it can be limited, especially for detection of the primary tumor. Improved renal cell cancer detection with carbonic anhydrase IX (CAIX)-based antibody (¹²⁴I-girentuximab) and radioimmunotherapy targeting with ¹⁷⁷Lu-cG250 appear promising. Evaluation of renal injury by imaging renal perfusion and function with novel PET radiotracers include p-¹⁸F-fluorohippurate, hippurate m-cyano-p-¹⁸F-fluorohippurate, and rubidium-82 chloride (typically used for myocardial perfusion imaging). Renal receptor imaging of the renal renin-angiotensin system with a variety of selective PET radioligands is also becoming available for clinical translation.
Collapse
Affiliation(s)
- Steve Y Cho
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD
| | - Zsolt Szabo
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD.
| |
Collapse
|
29
|
Persson M, El Ali HH, Binderup T, Pfeifer A, Madsen J, Rasmussen P, Kjaer A. Dosimetry of 64Cu-DOTA-AE105, a PET tracer for uPAR imaging. Nucl Med Biol 2014; 41:290-5. [DOI: 10.1016/j.nucmedbio.2013.12.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 11/28/2013] [Accepted: 12/07/2013] [Indexed: 01/08/2023]
|