1
|
Li Z, Ruan Q, Jiang Y, Wang Q, Yin G, Feng J, Zhang J. Current Status and Perspectives of Novel Radiopharmaceuticals with Heterologous Dual-targeted Functions: 2013-2023. J Med Chem 2024; 67:21644-21670. [PMID: 39648432 DOI: 10.1021/acs.jmedchem.4c01608] [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: 12/10/2024]
Abstract
Radiotracers provide molecular- and cellular-level information in a noninvasive manner and have become important tools for precision medicine. In particular, the successful clinical application of radioligand therapeutic (RLT) has further strengthened the role of nuclear medicine in clinical treatment. The complicated microenvironment of the lesion has rendered traditional single-targeted radiopharmaceuticals incapable of fully meeting the requirements. The design and development of dual-targeted and multitargeted radiopharmaceuticals have rapidly emerged. In recent years, significant progress has been made in the development of heterologous dual-targeted radiopharmaceuticals. This perspective aims to provide a comprehensive overview of the recent progress in these heterologous dual-targeted radiopharmaceuticals, with a special focus on the design of ligand structures, pharmacological properties, and preclinical and clinical evaluation. Furthermore, future directions are discussed from this perspective.
Collapse
Affiliation(s)
- Zuojie Li
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Qing Ruan
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
- Key Laboratory of Beam Technology of the Ministry of Education, College of Physics and Astronomy, Beijing Normal University, Beijing, 100875, P. R. China
| | - Yuhao Jiang
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
- Key Laboratory of Beam Technology of the Ministry of Education, College of Physics and Astronomy, Beijing Normal University, Beijing, 100875, P. R. China
| | - Qianna Wang
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Guangxing Yin
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Junhong Feng
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Junbo Zhang
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| |
Collapse
|
2
|
Sallam M, Nguyen NT, Sainsbury F, Kimizuka N, Muyldermans S, Benešová-Schäfer M. PSMA-targeted radiotheranostics in modern nuclear medicine: then, now, and what of the future? Theranostics 2024; 14:3043-3079. [PMID: 38855174 PMCID: PMC11155394 DOI: 10.7150/thno.92612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 04/04/2024] [Indexed: 06/11/2024] Open
Abstract
In 1853, the perception of prostate cancer (PCa) as a rare ailment prevailed, was described by the eminent Londoner surgeon John Adams. Rapidly forward to 2018, the landscape dramatically altered. Currently, men face a one-in-nine lifetime risk of PCa, accentuated by improved diagnostic methods and an ageing population. With more than three million men in the United States alone grappling with this disease, the overall risk of succumbing to stands at one in 39. The intricate clinical and biological diversity of PCa poses serious challenges in terms of imaging, ongoing monitoring, and disease management. In the field of theranostics, diagnostic and therapeutic approaches that harmoniously merge targeted imaging with treatments are integrated. A pivotal player in this arena is radiotheranostics, employing radionuclides for both imaging and therapy, with prostate-specific membrane antigen (PSMA) at the forefront. Clinical milestones have been reached, including FDA- and/or EMA-approved PSMA-targeted radiodiagnostic agents, such as [18F]DCFPyL (PYLARIFY®, Lantheus Holdings), [18F]rhPSMA-7.3 (POSLUMA®, Blue Earth Diagnostics) and [68Ga]Ga-PSMA-11 (Locametz®, Novartis/ ILLUCCIX®, Telix Pharmaceuticals), as well as PSMA-targeted radiotherapeutic agents, such as [177Lu]Lu-PSMA-617 (Pluvicto®, Novartis). Concurrently, ligand-drug and immune therapies designed to target PSMA are being advanced through rigorous preclinical research and clinical trials. This review delves into the annals of PSMA-targeted radiotheranostics, exploring its historical evolution as a signature molecule in PCa management. We scrutinise its clinical ramifications, acknowledge its limitations, and peer into the avenues that need further exploration. In the crucible of scientific inquiry, we aim to illuminate the path toward a future where the enigma of PCa is deciphered and where its menace is met with precise and effective countermeasures. In the following sections, we discuss the intriguing terrain of PCa radiotheranostics through the lens of PSMA, with the fervent hope of advancing our understanding and enhancing clinical practice.
Collapse
Affiliation(s)
- Mohamed Sallam
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- School of Environment and Science (ESC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- Griffith Institute for Drug Discovery (GRIDD), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Frank Sainsbury
- School of Environment and Science (ESC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- Griffith Institute for Drug Discovery (GRIDD), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Nobuo Kimizuka
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka 819-0395, Japan
- Center for Molecular Systems (CMS), Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka 819-0395, Japan
- Research Center for Negative Emissions Technologies (K-NETs), Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Martina Benešová-Schäfer
- Research Group Molecular Biology of Systemic Radiotherapy, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|
3
|
Jiao J, Zhang J, Wen W, Qin W, Chen X. Prostate-specific membrane antigen-targeted surgery in prostate cancer: Accurate identification, real-time diagnosis, and precise resection. Theranostics 2024; 14:2736-2756. [PMID: 38773975 PMCID: PMC11103491 DOI: 10.7150/thno.95039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 04/11/2024] [Indexed: 05/24/2024] Open
Abstract
Radical prostatectomy (RP) combined with pelvic lymph node dissection (PLND) is the first step in multimodal treatment of prostate cancer (PCa) without distant metastases. For a long time, the surgical resection range has been highly dependent on the surgeon's visualization and experience with preoperative imaging. With the rapid development of prostate-specific membrane antigen positron emission tomography and single-photon emission computed tomography (PSMA-PET and PSMA-SPECT), PSMA-targeted surgery has been introduced for a more accurate pathological diagnosis and complete resection of positive surgical margins (PSMs) and micro-lymph node metastases (LNMs). We reviewed PSMA-targeted surgeries, including PSMA-PET-guided prostatic biopsy (PSMA-TB), PSMA-targeted radio-guided surgery (PSMA-RGS), PSMA-targeted fluorescence-guided surgery (PSMA-FGS), and multi-modality/multi-targeted PSMA-targeted surgery. We also discuss the strengths and challenges of PSMA-targeted surgery, and propose that PSMA-targeted surgery could be a great addition to existing surgery protocols, thereby improving the accuracy and convenience of surgery for primary and recurrent PCa in the near future.
Collapse
Affiliation(s)
- Jianhua Jiao
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
- Innovation Center for Tumor Immunocytology Therapy Technology, Xijing Innovation Research Institute, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jingjing Zhang
- Departments of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Theranostics Center of Excellenece, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
| | - Weihong Wen
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
- Innovation Center for Tumor Immunocytology Therapy Technology, Xijing Innovation Research Institute, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore, Singapore
- Theranostics Center of Excellenece, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
| |
Collapse
|
4
|
Chambers C, Chitwood B, Smith CJ, Miao Y. Elevating theranostics: The emergence and promise of radiopharmaceutical cell-targeting heterodimers in human cancers. IRADIOLOGY 2024; 2:128-155. [PMID: 38708130 PMCID: PMC11067702 DOI: 10.1002/ird3.62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 01/30/2024] [Indexed: 05/07/2024]
Abstract
Optimal therapeutic and diagnostic efficacy is essential for healthcare's global mission of advancing oncologic drug development. Accurate diagnosis and detection are crucial prerequisites for effective risk stratification and personalized patient care in clinical oncology. A paradigm shift is emerging with the promise of multi-receptor-targeting compounds. While existing detection and staging methods have demonstrated some success, the traditional approach of monotherapy is being reevaluated to enhance therapeutic effectiveness. Heterodimeric site-specific agents are a versatile solution by targeting two distinct biomarkers with a single theranostic agent. This review describes the innovation of dual-targeting compounds, examining their design strategies, therapeutic implications, and the promising path they present for addressing complex diseases.
Collapse
Affiliation(s)
- Claudia Chambers
- Molecular Imaging and Theranostics Center, Columbia, Missouri, USA
- Research Division, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri, USA
- Department of Chemistry, University of Missouri, Columbia, Missouri, USA
| | - Broc Chitwood
- Molecular Imaging and Theranostics Center, Columbia, Missouri, USA
| | - Charles J. Smith
- Molecular Imaging and Theranostics Center, Columbia, Missouri, USA
- Research Division, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri, USA
- Department of Radiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- University of Missouri Research Reactor Center, University of Missouri, Columbia, Missouri, USA
| | - Yubin Miao
- Department of Radiology, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
5
|
Martin KE, Mattocks JA, Śmiłowicz D, Aluicio-Sarduy E, Whetter JN, Engle JW, Cotruvo JA, Boros E. Radiolabeling and in vivo evaluation of lanmodulin with biomedically relevant lanthanide isotopes. RSC Chem Biol 2023; 4:414-421. [PMID: 37292057 PMCID: PMC10246553 DOI: 10.1039/d3cb00020f] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/04/2023] [Indexed: 06/10/2023] Open
Abstract
Short-lived, radioactive lanthanides comprise an emerging class of radioisotopes attractive for biomedical imaging and therapy applications. To deliver such isotopes to target tissues, they must be appended to entities that target antigens overexpressed on the target cell's surface. However, the thermally sensitive nature of biomolecule-derived targeting vectors requires the incorporation of these isotopes without the use of denaturing temperatures or extreme pH conditions; chelating systems that can capture large radioisotopes under mild conditions are therefore highly desirable. Herein, we demonstrate the successful radiolabeling of the lanthanide-binding protein, lanmodulin (LanM), with medicinally relevant radioisotopes: 177Lu, 132/135La and 89Zr. Radiolabeling of the endogenous metal-binding sites of LanM, as well exogenous labeling of a protein-appended chelator, was successfully conducted at 25 °C and pH 7 with radiochemical yields ranging from 20-82%. The corresponding radiolabeled constructs possess good formulation stability in pH 7 MOPS buffer over 24 hours (>98%) in the presence of 2 equivalents of natLa carrier. In vivo experiments with [177Lu]-LanM, [132/135La]-LanM, and a prostate cancer targeting-vector linked conjugate, [132/135La]-LanM-PSMA, reveal that endogenously labeled constructs produce bone uptake in vivo. Exogenous, chelator-tag mediated radiolabeling to produce [89Zr]-DFO-LanM enables further study of the protein's in vivo behavior, demonstrating low bone and liver uptake, and renal clearance of the protein itself. While these results indicate that additional stabilization of LanM is required, this study establishes precedence for the radiochemical labeling of LanM with medically relevant lanthanide radioisotopes.
Collapse
Affiliation(s)
- Kirsten E Martin
- Department of Chemistry, Stony Brook University, Stony Brook New York 11794 USA
| | - Joseph A Mattocks
- Department of Chemistry, The Pennsylvania State University, University Park Pennsylvania 16802 USA
| | - Dariusz Śmiłowicz
- Department of Chemistry, Stony Brook University, Stony Brook New York 11794 USA
| | - Eduardo Aluicio-Sarduy
- Department of Medical Physics, University of Wisconsin Madison Wisconsin 53705 USA
- Department of Radiology, University of Wisconsin Madison Wisconsin 53705 USA
| | - Jennifer N Whetter
- Department of Chemistry, Stony Brook University, Stony Brook New York 11794 USA
| | - Jonathan W Engle
- Department of Medical Physics, University of Wisconsin Madison Wisconsin 53705 USA
- Department of Radiology, University of Wisconsin Madison Wisconsin 53705 USA
| | - Joseph A Cotruvo
- Department of Chemistry, The Pennsylvania State University, University Park Pennsylvania 16802 USA
| | - Eszter Boros
- Department of Chemistry, Stony Brook University, Stony Brook New York 11794 USA
| |
Collapse
|
6
|
Yan Q, Zhong J, Liu Y, Peng S, Feng P, Zhong Y, Hu K. Synthesis and preclinical evaluation of a heterodimeric radioligand targeting fibroblast activation protein and integrin-α vβ 3. Eur J Med Chem 2023; 251:115279. [PMID: 36931125 DOI: 10.1016/j.ejmech.2023.115279] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/13/2023]
Abstract
Tumor progression is accompanied by intrinsic heterogeneity and different phenotypes, which implies a different expression of cell surface receptors. Fibroblast activation protein (FAP) and integrin αvβ3 are highly expressed in the cell surface of cancer-associated cells or cancer cells compared with normal cells. Therefore, a FAP/integrin αvβ3 bispecific heterodimer was developed for positron emission tomography (PET) diagnostic imaging and radiotherapy. The heterodimer DOTA-FAPI-RGD was labeled with the diagnostic radionuclide gallium-68 or the therapeutic radionuclide lutetium-177, with yields >80%, and high stability. The competitive displacement binding assay showed an IC50 = 6.8 ± 0.6 nM for DOTA-FAPI-RGD towards FAP and IC50 = 2.1 ± 0.4 nM towards integrin αvβ3. Radionuclide labeled DOTA-FAPI-RGD showed high specificity and rapid internalization into U87MG cells (FAP/αvβ3-positive) in vitro. Micro-PET and biodistribution studies of [68Ga]Ga-DOTA-FAPI-RGD in tumor-bearing mice demonstrated that a high and specific tumor uptake of the tracer and a fast body clearance, resulting in high contrast images. In addition to the imaging applications demonstrated in this study, the labeling of the heterodimeric ligand with the radionuclide lutetium-177 used in cancer treatment might allow the therapeutic application of this ligand.
Collapse
Affiliation(s)
- Qingsong Yan
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province, 510515, China; Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Jiawei Zhong
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province, 510515, China; Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Yang Liu
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province, 510515, China
| | - Simin Peng
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province, 510515, China
| | - Pengju Feng
- Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Yuhua Zhong
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China.
| | - Kongzhen Hu
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province, 510515, China.
| |
Collapse
|
7
|
Boinapally S, Lisok A, Lofland G, Minn I, Yan Y, Jiang Z, Shin MJ, Merino VF, Zheng L, Brayton C, Pomper MG, Banerjee SR. Hetero-bivalent agents targeting FAP and PSMA. Eur J Nucl Med Mol Imaging 2022; 49:4369-4381. [PMID: 35965291 DOI: 10.1007/s00259-022-05933-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 08/01/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE We developed a theranostic radiopharmaceutical that engages two key cell surface proteases, fibroblast activation protein alpha (FAP) and prostate-specific membrane antigen (PSMA), each frequently overexpressed within the tumor microenvironment (TME). The latter is also expressed in most prostate tumor epithelium. To engage a broader spectrum of cancers for imaging and therapy, we conjugated small-molecule FAP and PSMA-targeting moieties using an optimized linker to provide 64Cu-labeled compounds. METHODS We synthesized FP-L1 and FP-L2 using two linker constructs attaching the FAP and PSMA-binding pharmacophores. We determined in vitro inhibition constants (Ki) for FAP and PSMA. Cell uptake assays and flow cytometry were conducted in human glioma (U87), melanoma (SK-MEL-24), prostate cancer (PSMA + PC3 PIP and PSMA - PC3 flu), and clear cell renal cell carcinoma lines (PSMA + /PSMA - 786-O). Quantitative positron emission tomography/computed tomography (PET/CT) and tissue biodistribution studies were performed using U87, SK-MEL-24, PSMA + PC3 PIP, and PSMA + 786-O experimental xenograft models and the KPC genetically engineered mouse model of pancreatic cancer. RESULTS 64Cu-FP-L1 and 64Cu-FP-L2 were produced in high radiochemical yields (> 98%) and molar activities (> 19 MBq/nmol). Ki values were in the nanomolar range for both FAP and PSMA. PET imaging and biodistribution studies revealed high and specific targeting of 64Cu-FP-L1 and 64Cu-FP-L2 for FAP and PSMA. 64Cu-FP-L1 displayed more favorable pharmacokinetics than 64Cu-FP-L2. In the U87 tumor model at 2 h post-injection, tumor uptake of 64Cu-FP-L1 (10.83 ± 1.02%ID/g) was comparable to 64Cu-FAPI-04 (9.53 ± 2.55%ID/g). 64Cu-FP-L1 demonstrated high retention 5.34 ± 0.29%ID/g at 48 h in U87 tumor. Additionally, 64Cu-FP-L1 showed high retention in PSMA + PC3 PIP tumor (12.06 ± 0.78%ID/g at 2 h and 10.51 ± 1.82%ID/g at 24 h). CONCLUSIONS 64Cu-FP-L1 demonstrated high and specific tumor targeting of FAP and PSMA. This compound should enable imaging of lesions expressing FAP, PSMA, or both on the tumor cell surface or within the TME. FP-L1 can readily be converted into a theranostic for the management of heterogeneous tumors.
Collapse
Affiliation(s)
- Srikanth Boinapally
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA
| | - Alla Lisok
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA
| | - Gabriela Lofland
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA
| | - Il Minn
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA
| | - Yu Yan
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA
| | - Zirui Jiang
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA
| | - Min Jay Shin
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA
| | - Vanessa F Merino
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA
| | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Cory Brayton
- Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA. .,Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Sangeeta Ray Banerjee
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA. .,Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| |
Collapse
|
8
|
Brunello S, Salvarese N, Carpanese D, Gobbi C, Melendez-Alafort L, Bolzati C. A Review on the Current State and Future Perspectives of [ 99mTc]Tc-Housed PSMA-i in Prostate Cancer. Molecules 2022; 27:molecules27092617. [PMID: 35565970 PMCID: PMC9099988 DOI: 10.3390/molecules27092617] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/12/2022] [Accepted: 04/12/2022] [Indexed: 02/01/2023] Open
Abstract
Recently, prostate-specific membrane antigen (PSMA) has gained momentum in tumor nuclear molecular imaging as an excellent target for both the diagnosis and therapy of prostate cancer. Since 2008, after years of preclinical research efforts, a plentitude of radiolabeled compounds mainly based on low molecular weight PSMA inhibitors (PSMA-i) have been described for imaging and theranostic applications, and some of them have been transferred to the clinic. Most of these compounds include radiometals (e.g., 68Ga, 64Cu, 177Lu) for positron emission tomography (PET) imaging or endoradiotherapy. Nowadays, although the development of new PET tracers has caused a significant drop in single-photon emission tomography (SPECT) research programs and the development of new technetium-99m (99mTc) tracers is rare, this radionuclide remains the best atom for SPECT imaging owing to its ideal physical decay properties, convenient availability, and rich and versatile coordination chemistry. Indeed, 99mTc still plays a relevant role in diagnostic nuclear medicine, as the number of clinical examinations based on 99mTc outscores that of PET agents and 99mTc-PSMA SPECT/CT may be a cost-effective alternative for 68Ga-PSMA PET/CT. This review aims to give an overview of the specific features of the developed [99mTc]Tc-tagged PSMA agents with particular attention to [99mTc]Tc-PSMA-i. The chemical and pharmacological properties of the latter will be compared and discussed, highlighting the pros and cons with respect to [68Ga]Ga-PSMA11.
Collapse
Affiliation(s)
- Sara Brunello
- Institute of Condensed Matter Chemistry and Technologies for Energy ICMATE-CNR, Corso Stati Uniti 4, 35127 Padova, Italy; (S.B.); (N.S.)
| | - Nicola Salvarese
- Institute of Condensed Matter Chemistry and Technologies for Energy ICMATE-CNR, Corso Stati Uniti 4, 35127 Padova, Italy; (S.B.); (N.S.)
| | - Debora Carpanese
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35124 Padova, Italy;
| | - Carolina Gobbi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy;
| | - Laura Melendez-Alafort
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35124 Padova, Italy;
- Correspondence: (L.M.-A.); (C.B.)
| | - Cristina Bolzati
- Institute of Condensed Matter Chemistry and Technologies for Energy ICMATE-CNR, Corso Stati Uniti 4, 35127 Padova, Italy; (S.B.); (N.S.)
- Correspondence: (L.M.-A.); (C.B.)
| |
Collapse
|
9
|
Neels OC, Kopka K, Liolios C, Afshar-Oromieh A. Radiolabeled PSMA Inhibitors. Cancers (Basel) 2021; 13:6255. [PMID: 34944875 PMCID: PMC8699044 DOI: 10.3390/cancers13246255] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 12/16/2022] Open
Abstract
PSMA has shown to be a promising target for diagnosis and therapy (theranostics) of prostate cancer. We have reviewed developments in the field of radio- and fluorescence-guided surgery and targeted photodynamic therapy as well as multitargeting PSMA inhibitors also addressing albumin, GRPr and integrin αvβ3. An overview of the regulatory status of PSMA-targeting radiopharmaceuticals in the USA and Europe is also provided. Technical and quality aspects of PSMA-targeting radiopharmaceuticals are described and new emerging radiolabeling strategies are discussed. Furthermore, insights are given into the production, application and potential of alternatives beyond the commonly used radionuclides for radiolabeling PSMA inhibitors. An additional refinement of radiopharmaceuticals is required in order to further improve dose-limiting factors, such as nephrotoxicity and salivary gland uptake during endoradiotherapy. The improvement of patient treatment achieved by the advantageous combination of radionuclide therapy with alternative therapies is also a special focus of this review.
Collapse
Affiliation(s)
- Oliver C. Neels
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstrasse 400, 01328 Dresden, Germany;
| | - Klaus Kopka
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstrasse 400, 01328 Dresden, Germany;
- Faculty of Chemistry and Food Chemistry, School of Science, Technical University Dresden, Mommsenstrasse 4, 01062 Dresden, Germany
| | - Christos Liolios
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, National & Kapodistrian University of Athens, Zografou, 15771 Athens, Greece;
- INRASTES, Radiochemistry Laboratory, NCSR “Demokritos”, Ag. Paraskevi Attikis, 15310 Athens, Greece
| | - Ali Afshar-Oromieh
- Department of Nuclear Medicine, Bern University Hospital (Inselspital), Freiburgstrasse 18, 3010 Bern, Switzerland;
| |
Collapse
|
10
|
Petrov SA, Zyk NY, Machulkin AE, Beloglazkina EK, Majouga AG. PSMA-targeted low-molecular double conjugates for diagnostics and therapy. Eur J Med Chem 2021; 225:113752. [PMID: 34464875 DOI: 10.1016/j.ejmech.2021.113752] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/27/2021] [Accepted: 08/05/2021] [Indexed: 12/24/2022]
Abstract
This review presents data on dual conjugates of therapeutic and diagnostic action for targeted delivery to prostate cancer cells. The works of the last ten years on this topic were analyzed. The mail attention focuses on low-molecular-weight conjugates directed to the prostate-specific membrane antigen (PSMA); the comparison of high and low molecular weight PSMA-targeted conjugates was made. The considered conjugates were divided in the review into two main classes: diagnostic bimodal conjugates (which are containing two fragments for different types of diagnostics), theranostic conjugates (containing both therapeutic and diagnostic agents); also bimodal high molecular weight therapeutic conjugates containing two therapeutic agents are briefly discussed. The data of in vitro and in vivo studies for PSMA-targeted double conjugates available by the beginning of 2021 have been analyzed.
Collapse
Affiliation(s)
- Stanislav A Petrov
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - Nikolay Y Zyk
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | | | | | - Alexander G Majouga
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia; Laboratory of Biomedical Nanomaterials, National University of Science and Technology MISiS, Moscow, Russia; Mendeleev University of Chemical Technology of Russia, Moscow, Russia
| |
Collapse
|
11
|
Zhang L, Shi X, Li Y, Duan X, Zhang Z, Fu H, Yang X, Tian J, Hu Z, Cui M. Visualizing Tumors in Real Time: A Highly Sensitive PSMA Probe for NIR-II Imaging and Intraoperative Tumor Resection. J Med Chem 2021; 64:7735-7745. [PMID: 34047189 DOI: 10.1021/acs.jmedchem.1c00444] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Owing to the complex anatomical structure, precise resection of a tumor while maintaining adjacent tissue is a challenge in radical prostatectomy for prostate cancer (PCa). Optical imaging in near-infrared window II (NIR-II) is a promising technology for intraoperative guidance, whereas there is no available probe for PCa yet. In this article, a novel probe (PSMA-1092) bearing two prostate-specific membrane antigen (PSMA) binding motifs was developed, displaying excellent optical properties (λmax = 1092 nm) and ultrahigh affinity (Ki = 80 pM) toward PSMA. The tumor was visualized with high resolution (tissue-to-normal tissue ratio = 7.62 ± 1.05) and clear margin by NIR-II imaging using PSMA-1092 in a mouse model. During the tumor resection, residual tumors missed by visible inspection were detected by the real-time imaging. Overall, PSMA-1092 displayed excellent performance in delineating the tumor margin and detecting residual tumors, demonstrating promising potential for precise PCa tumor resection in clinical practice.
Collapse
Affiliation(s)
- Longfei Zhang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Xiaojing Shi
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Yuying Li
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Xiaojiang Duan
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Zeyu Zhang
- School of Medical Science and Engineering, Beihang University, Beijing 100191, China
| | - Hualong Fu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Xing Yang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Zhenhua Hu
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, 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
|
12
|
Ha H, Kwon H, Lim T, Jang J, Park SK, Byun Y. Inhibitors of prostate-specific membrane antigen in the diagnosis and therapy of metastatic prostate cancer - a review of patent literature. Expert Opin Ther Pat 2021; 31:525-547. [PMID: 33459068 DOI: 10.1080/13543776.2021.1878145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Prostate-specific membrane antigen (PSMA), also known as glutamate carboxypeptidase II, is a potential target protein for imaging and treatment of patients with prostate cancer because of its overexpression during metastasis. Various PSMA-targeted imaging and therapeutic probes have been designed and synthesized based on the Lys-urea-Glu motif. Structural modifications have been made exclusively in the linker region, while maintaining the Lys-urea-Glu structure that interacts with S1 and S1' pockets. AREA COVERED This review includes WIPO-listed patents (from January 2017 to June 2020) reporting PSMA-targeted probes based on the Lys-urea-Glu or Glu-urea-Glu structure. EXPERT OPINION : PSMA-targeted imaging agents labeled with radionuclides such as fluorine-18, copper-64, gallium-68, and technetium-99m have been successfully translated into clinical phase for the early diagnosis of metastatic prostate cancer. Recently, PSMA-targeted therapeutic agents labeled with iodine-131, lutetium-177, astatine-211, and lead-212 have also been developed with notable progress. Most PSMA-targeted agents are based on the Lys-urea-Glu or Glu-urea-Glu structure, demonstrate strong PSMA-binding affinity in nanomolar range, and achieve diverse structural modifications in the non-pharmacophore pocket. By exploiting the S1 accessory pocket or the tunnel region of the PSMA active site, the in vivo efficacy and pharmacokinetic profiles of the PMSA-targeted agents can be effectively modulated.
Collapse
Affiliation(s)
- Hyunsoo Ha
- Department of Pharmacy, College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, South Korea
| | - Hongmok Kwon
- Department of Pharmacy, College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, South Korea
| | - Taehyeong Lim
- Department of Pharmacy, College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, South Korea
| | - Jaebong Jang
- Department of Pharmacy, College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, South Korea
| | - Song-Kyu Park
- Department of Pharmacy, College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, South Korea
| | - Youngjoo Byun
- Department of Pharmacy, College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, South Korea
| |
Collapse
|
13
|
Ludwig BS, Kessler H, Kossatz S, Reuning U. RGD-Binding Integrins Revisited: How Recently Discovered Functions and Novel Synthetic Ligands (Re-)Shape an Ever-Evolving Field. Cancers (Basel) 2021; 13:1711. [PMID: 33916607 PMCID: PMC8038522 DOI: 10.3390/cancers13071711] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 12/19/2022] Open
Abstract
Integrins have been extensively investigated as therapeutic targets over the last decades, which has been inspired by their multiple functions in cancer progression, metastasis, and angiogenesis as well as a continuously expanding number of other diseases, e.g., sepsis, fibrosis, and viral infections, possibly also Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2). Although integrin-targeted (cancer) therapy trials did not meet the high expectations yet, integrins are still valid and promising targets due to their elevated expression and surface accessibility on diseased cells. Thus, for the future successful clinical translation of integrin-targeted compounds, revisited and innovative treatment strategies have to be explored based on accumulated knowledge of integrin biology. For this, refined approaches are demanded aiming at alternative and improved preclinical models, optimized selectivity and pharmacological properties of integrin ligands, as well as more sophisticated treatment protocols considering dose fine-tuning of compounds. Moreover, integrin ligands exert high accuracy in disease monitoring as diagnostic molecular imaging tools, enabling patient selection for individualized integrin-targeted therapy. The present review comprehensively analyzes the state-of-the-art knowledge on the roles of RGD-binding integrin subtypes in cancer and non-cancerous diseases and outlines the latest achievements in the design and development of synthetic ligands and their application in biomedical, translational, and molecular imaging approaches. Indeed, substantial progress has already been made, including advanced ligand designs, numerous elaborated pre-clinical and first-in-human studies, while the discovery of novel applications for integrin ligands remains to be explored.
Collapse
Affiliation(s)
- Beatrice S. Ludwig
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
| | - Horst Kessler
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Susanne Kossatz
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Ute Reuning
- Clinical Research Unit, Department of Obstetrics and Gynecology, University Hospital Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany
| |
Collapse
|
14
|
Bandari RP, Carmack TL, Malhotra A, Watkinson L, Fergason Cantrell EA, Lewis MR, Smith CJ. Development of Heterobivalent Theranostic Probes Having High Affinity/Selectivity for the GRPR/PSMA. J Med Chem 2021; 64:2151-2166. [PMID: 33534560 DOI: 10.1021/acs.jmedchem.0c01785] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In this study, we describe the development of heterobivalent [DUPA-6-Ahx-([111In]In-DO3A)-8-Aoc-BBN ANT] and [DUPA-6-Ahx-([177Lu]Lu-DO3A)-8-Aoc-BBN ANT] radiotracers that display very high selectivity/specificity for gastrin-releasing peptide receptor (GRPR)-/prostate-specific membrane antigen (PSMA)-expressing cells. These studies include metallation, purification, characterization, and in vitro and in vivo evaluation of the new small-molecule-/peptide-based radiopharmaceuticals having utility for imaging and potentially therapy. Competitive displacement binding assays using PC-3 cells and LNCaP cell membranes showed high binding affinity for the GRPR or the PSMA. Biodistribution studies showed favorable excretion pharmacokinetics with high tumor uptake in PC-3 or PC-3 prostatic inhibin peptide (PIP) tumor-bearing mice. For example, tumor accumulation at the 1 h time point ranged from (4.74 ± 0.90) to (7.51 ± 2.61)%ID/g. Micro-single-photon emission computed tomography (microSPECT) molecular imaging investigations showed very high uptake in tumors with minimal accumulation of tracers in the surrounding collateral tissues in xenografted mice at 4 h postintravenous injection. In conclusion, [DUPA-6-Ahx-([111In]In-DO3A)-8-Aoc-BBN ANT] and [DUPA-6-Ahx-([177Lu]Lu-DO3A)-8-Aoc-BBN ANT] tracers displayed favorable pharmacokinetic and excretion profiles with high uptake and retention in tumors.
Collapse
Affiliation(s)
- Rajendra P Bandari
- Research Division, Harry S. Truman Memorial Veterans' Hospital, Research Service Room A005, 800 Hospital Drive, Columbia, Missouri 65201, United States.,Department of Radiology, University of Missouri School of Medicine, Columbia, Missouri 65211, United States
| | - Terry L Carmack
- Research Division, Harry S. Truman Memorial Veterans' Hospital, Research Service Room A005, 800 Hospital Drive, Columbia, Missouri 65201, United States.,University of Missouri Research Reactor Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Anil Malhotra
- Research Division, Harry S. Truman Memorial Veterans' Hospital, Research Service Room A005, 800 Hospital Drive, Columbia, Missouri 65201, United States
| | - Lisa Watkinson
- Research Division, Harry S. Truman Memorial Veterans' Hospital, Research Service Room A005, 800 Hospital Drive, Columbia, Missouri 65201, United States.,University of Missouri Research Reactor Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Emily A Fergason Cantrell
- Research Division, Harry S. Truman Memorial Veterans' Hospital, Research Service Room A005, 800 Hospital Drive, Columbia, Missouri 65201, United States.,University of Missouri Research Reactor Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Michael R Lewis
- Department of Veterinary Medicine and Surgery, University of Missouri College of Veterinary Medicine, Columbia, Missouri 65211, United States
| | - Charles J Smith
- Research Division, Harry S. Truman Memorial Veterans' Hospital, Research Service Room A005, 800 Hospital Drive, Columbia, Missouri 65201, United States.,Department of Radiology, University of Missouri School of Medicine, Columbia, Missouri 65211, United States.,University of Missouri Research Reactor Center, University of Missouri, Columbia, Missouri 65211, United States
| |
Collapse
|
15
|
Hensbergen A, van Willigen DM, van Beurden F, van Leeuwen PJ, Buckle T, Schottelius M, Maurer T, Wester HJ, van Leeuwen FWB. Image-Guided Surgery: Are We Getting the Most Out of Small-Molecule Prostate-Specific-Membrane-Antigen-Targeted Tracers? Bioconjug Chem 2020; 31:375-395. [PMID: 31855410 PMCID: PMC7033908 DOI: 10.1021/acs.bioconjchem.9b00758] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/19/2019] [Indexed: 12/12/2022]
Abstract
Expressed on virtually all prostate cancers and their metastases, the transmembrane protein prostate-specific membrane antigen (PSMA) provides a valuable target for the imaging of prostate cancer. Not only does PSMA provide a target for noninvasive diagnostic imaging, e.g., PSMA-positron emission tomography (PSMA-PET), it can also be used to guide surgical resections of PSMA-positive lesions. The latter characteristic has led to the development of a plethora of PSMA-targeted tracers, i.e., radiolabeled, fluorescent, or hybrid. With image-guided surgery applications in mind, this review discusses these compounds based on clinical need. Here, the focus is on the chemical aspects (e.g., imaging label, spacer moiety, and targeting vector) and their impact on in vitro and in vivo tracer characteristics (e.g., affinity, tumor uptake, and clearance pattern).
Collapse
Affiliation(s)
- Albertus
Wijnand Hensbergen
- Interventional
Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Danny M. van Willigen
- Interventional
Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Florian van Beurden
- Interventional
Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department
of Urology, Netherlands Cancer Institute-Antoni
van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands
| | - Pim J. van Leeuwen
- Department
of Urology, Netherlands Cancer Institute-Antoni
van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands
| | - Tessa Buckle
- Interventional
Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department
of Urology, Netherlands Cancer Institute-Antoni
van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands
| | - Margret Schottelius
- Translational
Radiopharmaceutical Sciences, Department of Nuclear Medicine, Centre
Hospitalier Universitaire Vaudois (CHUV) and Department of Oncology, University of Lausanne (UNIL), 1011 Lausanne, Switzerland
| | - Tobias Maurer
- Department
of Urology and Martini-Klinik, Universitätsklinikum
Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Hans-Jürgen Wester
- Pharmazeutische
Radiochemie, Technische Universität
München, 85748 Garching, Germany
| | - Fijs W. B. van Leeuwen
- Interventional
Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department
of Urology, Netherlands Cancer Institute-Antoni
van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
16
|
Ruigrok EAM, van Weerden WM, Nonnekens J, de Jong M. The Future of PSMA-Targeted Radionuclide Therapy: An Overview of Recent Preclinical Research. Pharmaceutics 2019; 11:E560. [PMID: 31671763 PMCID: PMC6921028 DOI: 10.3390/pharmaceutics11110560] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 12/15/2022] Open
Abstract
Prostate specific membrane antigen (PSMA) has become a major focus point in the research and development of prostate cancer (PCa) imaging and therapeutic strategies using radiolabeled tracers. PSMA has shown to be an excellent target for PCa theranostics because of its high expression on the membrane of PCa cells and the increase in expression during disease progression. Therefore, numerous PSMA-targeting tracers have been developed and (pre)clinically studied with promising results. However, many of these PSMA-targeting tracers show uptake in healthy organs such as the salivary glands, causing radiotoxicity. Furthermore, not all patients respond to PSMA-targeted radionuclide therapy (TRT). This created the necessity of additional preclinical research studies in which existing tracers are reevaluated and new tracers are developed in order to improve PSMA-TRT by protecting the (PSMA-expressing) healthy organs and improving tumor uptake. In this review we will give an overview of the recent preclinical research projects regarding PCa-TRT using PSMA-specific radiotracers, which will give an indication of where the PSMA-TRT research movement is going and what we can expect in future clinical trials.
Collapse
Affiliation(s)
- Eline A M Ruigrok
- Dept. of Radiology and Nuclear Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands.
- Dept. of Experimental Urology, Erasmus MC, 3015 GD Rotterdam, The Netherlands.
| | | | - Julie Nonnekens
- Dept. of Radiology and Nuclear Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands.
- Dept. of Molecular Genetics, Erasmus MC, 3015 GD Rotterdam, The Netherlands.
- Oncode Institute, Erasmus MC, 3015 GD Rotterdam, The Netherlands.
| | - Marion de Jong
- Dept. of Radiology and Nuclear Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands.
| |
Collapse
|
17
|
Kwon H, Son S, Byun Y. Prostate‐Specific Membrane Antigen (PSMA)‐Targeted Radionuclide Probes for Imaging and Therapy of Prostate Cancer. ASIAN J ORG CHEM 2019. [DOI: 10.1002/ajoc.201900329] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Hongmok Kwon
- College of PharmacyKorea University 2511 Sejong-ro Sejong 30019 South Korea
| | - Sang‐Hyun Son
- College of PharmacyKorea University 2511 Sejong-ro Sejong 30019 South Korea
| | - Youngjoo Byun
- College of PharmacyKorea University 2511 Sejong-ro Sejong 30019 South Korea
| |
Collapse
|
18
|
Wang L, Liang H, Sun J, Liu Y, Li J, Li J, Li J, Yang H. Bispecific Aptamer Induced Artificial Protein-Pairing: A Strategy for Selective Inhibition of Receptor Function. J Am Chem Soc 2019; 141:12673-12681. [PMID: 31381313 DOI: 10.1021/jacs.9b05123] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cell surface receptors play a critical role in modulating intracellular signal transduction, making them important drug targets. However, it remains challenging to develop a selective and efficient strategy for regulating receptor function. Herein, we develop a strategy, called bispecific aptamer induced artificial protein-pairing, to selectively regulate receptor function. In this strategy, bispecific aptamer probes act as molecular mediators to bind to both a target receptor protein and a paired protein, which brings the two proteins into close proximity on the living cell membrane. Importantly, the paired proteins work not only as a cancer biomarker for enhancing cell selectivity but also as a blocking assistant to inhibit target receptor function via strong steric hindrance effect. Compared with single-aptamer-mediated regulation, the proposed bispecific aptamer probes afford substantial improvement in selective and efficient regulation of receptor function and downstream signaling pathways. This work offers a versatile methodology to design molecular mediators that can modulate receptor function, thereby providing a new way for developing novel therapeutic drugs.
Collapse
Affiliation(s)
- Liping Wang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry , Fuzhou University , Fuzhou 350108 , People's Republic of China.,Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University , Shanghai 200240 , People's Republic of China
| | - Hong Liang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry , Fuzhou University , Fuzhou 350108 , People's Republic of China.,Institute of Oceanography , Minjiang University , Fuzhou , Fujian 350108 , People's Republic of China
| | - Jin Sun
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University , Shanghai 200240 , People's Republic of China.,College of Biological Science and Engineering , Fuzhou University , Fuzhou 350108 , People's Republic of China
| | - Yichang Liu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry , Fuzhou University , Fuzhou 350108 , People's Republic of China
| | - Jinyu Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry , Fuzhou University , Fuzhou 350108 , People's Republic of China
| | - Jingying Li
- College of Biological Science and Engineering , Fuzhou University , Fuzhou 350108 , People's Republic of China
| | - Juan Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry , Fuzhou University , Fuzhou 350108 , People's Republic of China.,Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University , Shanghai 200240 , People's Republic of China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry , Fuzhou University , Fuzhou 350108 , People's Republic of China
| |
Collapse
|
19
|
Liolios C, Sachpekidis C, Schäfer M, Kopka K. Bispecific radioligands targeting prostate-specific membrane antigen and gastrin-releasing peptide receptors on the surface of prostate cancer cells. J Labelled Comp Radiopharm 2019; 62:510-522. [DOI: 10.1002/jlcr.3749] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/15/2019] [Accepted: 05/03/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Christos Liolios
- Division of Radiopharmaceutical Chemistry; German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
| | - Christos Sachpekidis
- Clinical Cooperation Unit Nuclear Medicine; German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
| | - Martin Schäfer
- Division of Radiopharmaceutical Chemistry; German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
| | - Klaus Kopka
- Division of Radiopharmaceutical Chemistry; German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
- German Cancer Consortium (DKTK); German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
| |
Collapse
|
20
|
Evaluation of Lung Cancer and Neuroendocrine Neoplasm in a Single Scan by Targeting Both Somatostatin Receptor and Integrin αvβ3. Clin Nucl Med 2019; 44:687-694. [DOI: 10.1097/rlu.0000000000002680] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
21
|
Ivanenkov YA, Machulkin AE, Garanina AS, Skvortsov DA, Uspenskaya AA, Deyneka EV, Trofimenko AV, Beloglazkina EK, Zyk NV, Koteliansky VE, Bezrukov DS, Aladinskaya AV, Vorobyeva NS, Puchinina MM, Riabykh GK, Sofronova AA, Malyshev AS, Majouga AG. Synthesis and biological evaluation of Doxorubicin-containing conjugate targeting PSMA. Bioorg Med Chem Lett 2019; 29:1246-1255. [DOI: 10.1016/j.bmcl.2019.01.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/15/2019] [Accepted: 01/30/2019] [Indexed: 12/19/2022]
|
22
|
Chen J, Gao Z, Li G, Wang TD. Dual-modal in vivo fluorescence and photoacoustic imaging using a heterodimeric peptide. Chem Commun (Camb) 2018; 54:13196-13199. [PMID: 30334022 DOI: 10.1039/c8cc06774k] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
A heterodimeric peptide labeled with IRDye800 is used to perform dual-modal imaging of human esophageal xenograft tumors in vivo. Fluorescence and photoacoustic images provide complementary visualization of tumor dimensions in planar and sagittal views, respectively, demonstrating promise for targeted cancer diagnosis and staging.
Collapse
Affiliation(s)
- Jing Chen
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | |
Collapse
|
23
|
Tian JY, Guo FJ, Zheng GY, Ahmad A. Prostate cancer: updates on current strategies for screening, diagnosis and clinical implications of treatment modalities. Carcinogenesis 2018; 39:307-317. [PMID: 29216344 DOI: 10.1093/carcin/bgx141] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 11/29/2017] [Indexed: 01/23/2023] Open
Abstract
Prostate cancer is the most common cancer in men by way of diagnosis and a leading cause of cancer-related deaths. Early detection and intervention remains key to its optimum clinical management. This review provides the most updated information on the recent methods of prostate cancer screening, imaging and treatment modalities. Wherever possible, clinical trial data has been supplemented to provide a comprehensive overview of current prostate cancer research and development. Considering the recent success of immunotherapy in prostate cancer, we discuss cell, DNA and viruses based, as well as combinatorial immunotherapeutic strategies in detail. Furthermore, the potential of nanotechnology is increasingly being realized, especially in prostate cancer research, and we provide an overview of nanotechnology-based strategies, with special emphasis on nanotheranostics and multifunctional nanoconstructs. Understanding these recent developments is critical to the design of future therapeutic strategies to counter prostate cancer.
Collapse
Affiliation(s)
- Jing-Yan Tian
- Department of Urology, Second Division of the First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Feng-Jun Guo
- Department of Gynaecology and Obstetrics, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Guo-You Zheng
- Department of Urology, Second Division of the First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Aamir Ahmad
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| |
Collapse
|
24
|
Wang D, Sun Y, Liu Y, Meng F, Lee RJ. Clinical translation of immunoliposomes for cancer therapy: recent perspectives. Expert Opin Drug Deliv 2018; 15:893-903. [PMID: 30169978 DOI: 10.1080/17425247.2018.1517747] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Liposomes have been extensively investigated as drug delivery vehicles. Immunoliposomes (ILs) are antibody-conjugated liposomes designed to selectively target antigen-expressing cells. ILs can be used to deliver drugs to tumor cells for improving efficacy and reducing toxicity. In addition, ILs can be used in immunoassays, immunotherapy, and imaging. Although there has been extensive coverage on ILs in the literature, only a limited number of clinical trials have been reported and no IL drug has been approved by the FDA. AREAS COVERED Factors to consider in developing ILs are discussed, including the choice of antibody or antibody fragment, the formulation of liposomes, and the conjugation chemistry. In addition, challenges and opportunities in clinical development of ILs are discussed. The purpose of this review is to provide an overview on the state of the art of ILs and to discuss potential future developments. EXPERT OPINION IL research has had a lengthy history and numerous preclinical studies have yielded encouraging results. However, there are a number of obstacles to clinical translation of ILs. Given the unique capabilities of ILs, its potential for clinical application is underexplored. There is great potential for expanded role for ILs in the clinic and further efforts to this end are warranted. ABBREVIATIONS Ab: antibody; ADCs: antibody-drug conjugates; API: active pharmaceutical ingredient; ADCC: antibody-dependent cellular cytotoxicity; CR: complete remission; cGMP: current good manufacturing practice; DSPE: distearoyl phosphatidylethanolamine; EGF: epidermal growth factor; EGFR: epidermal growth factor receptor; EPR: enhanced permeability and retention; Fc: fragment crystalline; Tf: transferrin; HACA: human-anti-chimeric antibody; HAHA: human-anti-human antibody; HAMA: human-anti-mouse antibody; HER2: human epidermal growth factor 2; IL: immunoliposome; LNPs: lipid nanoparticles; MRI: magnetic resonance imaging; MTD: maximum tolerated dose; PEG: polyethylene glycol; PET: positron emission tomography; PR: partial response; PSMA: prostate-specific membrane antigen; scFv: single-chain variable fragment; SPECT: single photon emission computed tomography; TTR: transthyretin.
Collapse
Affiliation(s)
- Di Wang
- a School of Life Sciences , Jilin University , Changchun , China
| | - Yating Sun
- a School of Life Sciences , Jilin University , Changchun , China
| | - Yange Liu
- a School of Life Sciences , Jilin University , Changchun , China
| | - Fanchao Meng
- a School of Life Sciences , Jilin University , Changchun , China
| | - Robert J Lee
- a School of Life Sciences , Jilin University , Changchun , China.,b Division of Pharmaceutics and Pharmaceutical Chemistry , The Ohio State University , Columbus , OH , USA
| |
Collapse
|
25
|
Chang CA, Chia J, Lin S. A Cyclic‐RGD Dinuclear Tb
III
Macrocyclic Complex as a Tumor Integrin‐Selective Luminescent Probe. Eur J Inorg Chem 2018. [DOI: 10.1002/ejic.201800568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- C. Allen Chang
- Department of Biomedical Imaging and Radiological Sciences National Yang‐Ming University No. 155, Sec. 2, Li‐Nong St., Beitou 112 Taipei R.O.C. Taiwan
- Department of Biotechnology and Laboratory Science in Medicine National Yang‐Ming University R.O.C. Taiwan
- Molecular Imaging Research Center National Yang‐Ming University R.O.C. Taiwan
| | - Ju‐Chien Chia
- Department of Biomedical Imaging and Radiological Sciences National Yang‐Ming University No. 155, Sec. 2, Li‐Nong St., Beitou 112 Taipei R.O.C. Taiwan
| | - Syue‐Liang Lin
- Department of Biotechnology and Laboratory Science in Medicine National Yang‐Ming University R.O.C. Taiwan
- Molecular Imaging Research Center National Yang‐Ming University R.O.C. Taiwan
| |
Collapse
|
26
|
Chen J, Zhou J, Gao Z, Li X, Wang F, Duan X, Li G, Joshi BP, Kuick R, Appelman HD, Wang TD. Multiplexed Targeting of Barrett's Neoplasia with a Heterobivalent Ligand: Imaging Study on Mouse Xenograft in Vivo and Human Specimens ex Vivo. J Med Chem 2018; 61:5323-5331. [PMID: 29870260 DOI: 10.1021/acs.jmedchem.8b00405] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Esophageal adenocarcinoma (EAC) is a molecularly heterogeneous disease that is rising rapidly in incidence and has poor prognosis. We developed a heterobivalent peptide to target detection of early Barrett's neoplasia by combining monomer heptapeptides specific for either EGFR or ErbB2 in a heterodimer configuration. The structure of a triethylene glycol linker was optimized to maximize binding interactions to the surface receptors on cells. The Cy5.5-labeled heterodimer QRH*-KSP*-E3-Cy5.5 demonstrated specific binding to each target and showed 3-fold greater fluorescence intensity and 2-fold higher affinity compared with those of either monomer alone. Peak uptake in xenograft tumors was observed at 2 h postinjection with systemic clearance by ∼24 h in vivo. Furthermore, ligand binding was evaluated on human esophageal specimens ex vivo, and 88% sensitivity and 87% specificity were found for the detection of either high-grade dysplasia (HGD) or EAC. This peptide heterodimer shows promise for targeted detection of early Barrett's neoplasia in clinical study.
Collapse
|
27
|
Peptide-Based Radiopharmaceuticals for Molecular Imaging of Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1096:135-158. [DOI: 10.1007/978-3-319-99286-0_8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
28
|
Preparation and in vitro evaluation of 177Lu-iPSMA-RGD as a new heterobivalent radiopharmaceutical. J Radioanal Nucl Chem 2017. [DOI: 10.1007/s10967-017-5555-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
29
|
González-Ruíz A, Ferro-Flores G, Azorín-Vega E, Ocampo-García B, Ramírez FDM, Santos-Cuevas C, De León-Rodríguez L, Isaac-Olivé K, Luna-Gutiérrez M, Morales-Ávila E. Synthesis and in vitro evaluation of an antiangiogenic cancer-specific dual-targeting 177Lu-Au-nanoradiopharmaceutical. J Radioanal Nucl Chem 2017. [DOI: 10.1007/s10967-017-5465-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
30
|
Reubi JC, Maecke HR. Approaches to Multireceptor Targeting: Hybrid Radioligands, Radioligand Cocktails, and Sequential Radioligand Applications. J Nucl Med 2017; 58:10S-16S. [DOI: 10.2967/jnumed.116.186882] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 07/19/2017] [Indexed: 01/21/2023] Open
|
31
|
Flores O, Santra S, Kaittanis C, Bassiouni R, Khaled AS, Khaled AR, Grimm J, Perez JM. PSMA-Targeted Theranostic Nanocarrier for Prostate Cancer. Am J Cancer Res 2017; 7:2477-2494. [PMID: 28744329 PMCID: PMC5525751 DOI: 10.7150/thno.18879] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 04/12/2017] [Indexed: 11/05/2022] Open
Abstract
Herein, we report the use of a theranostic nanocarrier (Folate-HBPE(CT20p)) to deliver a therapeutic peptide to prostate cancer tumors that express PSMA (folate hydrolase 1). The therapeutic peptide (CT20p) targets and inhibits the chaperonin-containing TCP-1 (CCT) protein-folding complex, is selectively cytotoxic to cancer cells, and is non-toxic to normal tissue. With the delivery of CT20p to prostate cancer cells via PSMA, a dual level of cancer specificity is achieved: (1) selective targeting to PSMA-expressing prostate tumors, and (2) specific cytotoxicity to cancer cells with minimal toxicity to normal cells. The PSMA-targeting theranostic nanocarrier can image PSMA-expressing cells and tumors when a near infrared dye is used as cargo. Meanwhile, it can be used to treat PSMA-expressing tumors when a therapeutic, such as the CT20p peptide, is encapsulated within the nanocarrier. Even when these PSMA-targeting nanocarriers are taken up by macrophages, minimal cell death is observed in these cells, in contrast with doxorubicin-based therapeutics that result in significant macrophage death. Incubation of PSMA-expressing prostate cancer cells with the Folate-HBPE(CT20p) nanocarriers induces considerable changes in cell morphology, reduction in the levels of integrin β1, and lower cell adhesion, eventually resulting in cell death. These results are relevant as integrin β1 plays a key role in prostate cancer invasion and metastatic potential. In addition, the use of the developed PSMA-targeting nanocarrier facilitates the selective in vivo delivery of CT20p to PSMA-positive tumor, inducing significant reduction in tumor size.
Collapse
|
32
|
Compounds for radionuclide imaging and therapy of malignant foci characterized by the increased angiogenesis. Russ Chem Bull 2016. [DOI: 10.1007/s11172-016-1309-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
33
|
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
|
34
|
Subedi M, Minn I, Chen J, Kim Y, Ok K, Jung YW, Pomper MG, Byun Y. Design, synthesis and biological evaluation of PSMA/hepsin-targeted heterobivalent ligands. Eur J Med Chem 2016; 118:208-218. [PMID: 27128184 DOI: 10.1016/j.ejmech.2016.04.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/14/2016] [Accepted: 04/12/2016] [Indexed: 01/08/2023]
Abstract
Cell surface biomarkers such as prostate-specific membrane antigen (PSMA) and hepsin have received considerable attention as targets for imaging prostate cancer (PCa) due to their high cell surface expression in such tumors and easy access for imaging probes. Novel amidine-containing indole analogs (13-21) as hepsin inhibitors were designed and synthesized. These compounds showed in vitro inhibitory activity against hepsin with IC50 values from 5.9 to 70 μM. Based on the SAR of amidine-derived analogs, the novel heterobivalent compound 30, targeting both hepsin and PSMA, was synthesized by linking compound 18 with Lys-urea-Glu, the key scaffold for the specific binding to PSMA, followed by the conjugation of the optical dye SulfoCy7. Compound 30 exhibited inhibitory activities against PSMA and hepsin, with IC50 values of 28 nM and 2.8 μM, respectively. In vitro cell uptake and preliminary in vivo optical imaging studies of 30 showed selective binding and retention in both PSMA and hepsin high-expressing PC3/ML-PSMA-HPN cells as compared with low-expressing PC3/ML cells.
Collapse
Affiliation(s)
- Milan Subedi
- College of Pharmacy, Korea University, 2511 Sejong-ro, Jochiwon-eup, Sejong 339-700, South Korea
| | - Il Minn
- Department of Radiology, Johns Hopkins Medical Institution, 1550 Orleans street, Baltimore 21287, MD, USA
| | - Jianbo Chen
- College of Pharmacy, Korea University, 2511 Sejong-ro, Jochiwon-eup, Sejong 339-700, South Korea
| | - YunHye Kim
- College of Pharmacy, Korea University, 2511 Sejong-ro, Jochiwon-eup, Sejong 339-700, South Korea
| | - Kiwon Ok
- College of Pharmacy, Korea University, 2511 Sejong-ro, Jochiwon-eup, Sejong 339-700, South Korea
| | - Yong Woo Jung
- College of Pharmacy, Korea University, 2511 Sejong-ro, Jochiwon-eup, Sejong 339-700, South Korea
| | - Martin G Pomper
- Department of Radiology, Johns Hopkins Medical Institution, 1550 Orleans street, Baltimore 21287, MD, USA
| | - Youngjoo Byun
- College of Pharmacy, Korea University, 2511 Sejong-ro, Jochiwon-eup, Sejong 339-700, South Korea.,Department of Radiology, Johns Hopkins Medical Institution, 1550 Orleans street, Baltimore 21287, MD, USA.,Biomedical Research Center, Korea University Guro Hospital, 148 Gurodong-ro, Guro-gu, Seoul 152-703, South Korea
| |
Collapse
|
35
|
Williford JM, Archang MM, Minn I, Ren Y, Wo M, Vandermark J, Fisher PB, Pomper MG, Mao HQ. Critical Length of PEG Grafts on lPEI/DNA Nanoparticles for Efficient in Vivo Delivery. ACS Biomater Sci Eng 2016; 2:567-578. [PMID: 27088129 PMCID: PMC4829937 DOI: 10.1021/acsbiomaterials.5b00551] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/02/2016] [Indexed: 12/03/2022]
Abstract
![]()
Nanoparticle-mediated
gene delivery is a promising alternative
to viral methods; however, its use in vivo, particularly following
systemic injection, has suffered from poor delivery efficiency. Although
PEGylation of nanoparticles has been successfully demonstrated as
a strategy to enhance colloidal stability, its success in improving
delivery efficiency has been limited, largely due to reduced cell
binding and uptake, leading to poor transfection efficiency. Here
we identified an optimized PEGylation scheme for DNA micellar nanoparticles
that delivers balanced colloidal stability and transfection activity.
Using linear polyethylenimine (lPEI)-g-PEG as a carrier,
we characterized the effect of graft length and density of polyethylene
glycol (PEG) on nanoparticle assembly, micelle stability, and gene
delivery efficiency. Through variation of PEG grafting degree, lPEI
with short PEG grafts (molecular weight, MW 500–700 Da) generated
micellar nanoparticles with various shapes including spherical, rodlike,
and wormlike nanoparticles. DNA micellar nanoparticles prepared with
short PEG grafts showed comparable colloidal stability in salt and
serum-containing media to those prepared with longer PEG grafts (MW
2 kDa). Corresponding to this trend, nanoparticles prepared with short
PEG grafts displayed significantly higher in vitro transfection efficiency
compared to those with longer PEG grafts. More importantly, short
PEG grafts permitted marked increase in transfection efficiency following
ligand conjugation to the PEG terminal in metastatic prostate cancer-bearing
mice. This study identifies that lPEI-g-PEG with
short PEG grafts (MW 500–700 Da) is the most effective to ensure
shape control and deliver high colloidal stability, transfection activity,
and ligand effect for DNA nanoparticles in vitro and in vivo following
intravenous administration.
Collapse
Affiliation(s)
- John-Michael Williford
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, Maryland 21205, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States
| | - Maani M Archang
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States
| | - Il Minn
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins Medical Institutions , 601 N. Caroline Street, Baltimore, Maryland 21287, United States
| | - Yong Ren
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States
| | - Mark Wo
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States
| | - John Vandermark
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, 1101 East Marshall Street, Richmond, Virginia 23298, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, 1220 East Broad Street, Richmond, Virginia 23298, United States; VCU Massey Cancer Center, Virginia Commonwealth University, 401 College Street, Richmond, Virginia 23298, United States
| | - Martin G Pomper
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins Medical Institutions, 601 N. Caroline Street, Baltimore, Maryland 21287, United States
| | - Hai-Quan Mao
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Translational Tissue Engineering Center and Whitaker Biomedical Engineering Institute, Johns Hopkins University School of Medicine, 400 N. Broadway, Baltimore, Maryland 21287, United States
| |
Collapse
|
36
|
Liolios C, Schäfer M, Haberkorn U, Eder M, Kopka K. Novel Bispecific PSMA/GRPr Targeting Radioligands with Optimized Pharmacokinetics for Improved PET Imaging of Prostate Cancer. Bioconjug Chem 2016; 27:737-51. [PMID: 26726823 DOI: 10.1021/acs.bioconjchem.5b00687] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A new series of bispecific radioligands (BRLs) targeting prostate-specific membrane antigen (PSMA) and gastrin releasing peptide receptor (GRPr), both expressed on prostate cancer cells, was developed. Their design was based on the bombesin (BN) analogue, H2N-PEG2-[D-Tyr(6),β-Ala(11),Thi(13),Nle(14)]BN(6-14), which binds to GRPr with high affinity and specificity, and the peptidomimetic urea-based pseudoirreversible inhibitor of PSMA, Glu-ureido-Lys. The two pharmacophores were coupled through copper(I)-catalyzed azide-alkyne cycloaddition to the bis(tetrafluorophenyl) ester of the chelating agent HBED-CC via amino acid linkers made of positively charged His (H) and negatively charged Glu (E): -(HE)n- (n = 0-3). The BRLs were labeled with (68)Ga, and their preliminary pharmacological properties were evaluated in vitro (competitive and time kinetic binding assays) on prostate cancer (PC-3, LNCaP) and rat pancreatic (AR42J) cell lines and in vivo by biodistribution and small animal PET imaging studies in both normal and tumor-bearing mice. The IC50/Ki values determined for all BRLs essentially matched those of the respective monomers. The maximal cellular uptake of the BLRs was observed between 20 and 30 min. The BRLs showed a synergistic ability in vivo by targeting both PSMA (LNCaP) and GRPr (PC-3) positive tumors, whereas the charged -(HE)n- (n = 1-3) linkers significantly reduced the kidney and spleen uptake. The bispecific (PSMA and GRPr) targeting ability and optimized pharmacokinetics of the compounds developed in this study could lead to their future application in clinical practice as more sensitive radiotracers for noninvasive imaging of prostate cancer (PCa) by PET/CT and PET/MRI.
Collapse
Affiliation(s)
- C Liolios
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ) , 69120 Heidelberg, Germany
| | - M Schäfer
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ) , 69120 Heidelberg, Germany
| | - U Haberkorn
- Clinical Cooperation Unit Nuclear Medicine, University of Heidelberg , 69120 Heidelberg, Germany.,German Cancer Consortium (DKTK) , 69120 Heidelberg, Germany
| | - M Eder
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ) , 69120 Heidelberg, Germany
| | - K Kopka
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ) , 69120 Heidelberg, Germany.,German Cancer Consortium (DKTK) , 69120 Heidelberg, Germany
| |
Collapse
|
37
|
Kiess AP, Banerjee SR, Mease RC, Rowe SP, Rao A, Foss CA, Chen Y, Yang X, Cho SY, Nimmagadda S, Pomper MG. Prostate-specific membrane antigen as a target for cancer imaging and therapy. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2015; 59:241-68. [PMID: 26213140 PMCID: PMC4859214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
The prostate-specific membrane antigen (PSMA) is a molecular target whose use has resulted in some of the most productive work toward imaging and treating prostate cancer over the past two decades. A wide variety of imaging agents extending from intact antibodies to low-molecular-weight compounds permeate the literature. In parallel there is a rapidly expanding pool of antibody-drug conjugates, radiopharmaceutical therapeutics, small-molecule drug conjugates, theranostics and nanomedicines targeting PSMA. Such productivity is motivated by the abundant expression of PSMA on the surface of prostate cancer cells and within the neovasculature of other solid tumors, with limited expression in most normal tissues. Animating the field is a variety of small-molecule scaffolds upon which the radionuclides, drugs, MR-detectable species and nanoparticles can be placed with relative ease. Among those, the urea-based agents have been most extensively leveraged, with expanding clinical use for detection and more recently for radiopharmaceutical therapy of prostate cancer, with surprisingly little toxicity. PSMA imaging of other cancers is also appearing in the clinical literature, and may overtake FDG for certain indications. Targeting PSMA may provide a viable alternative or first-line approach to managing prostate and other cancers.
Collapse
Affiliation(s)
- A P Kiess
- Department of Radiation Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, USA -
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Nani RR, Shaum JB, Gorka AP, Schnermann MJ. Electrophile-integrating Smiles rearrangement provides previously inaccessible C4'-O-alkyl heptamethine cyanine fluorophores. Org Lett 2015; 17:302-5. [PMID: 25562683 PMCID: PMC4301176 DOI: 10.1021/ol503398f] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
![]()
New synthetic methods to rapidly
access useful fluorophores are
needed to advance modern molecular imaging techniques. A new variant
of the classical Smiles rearrangement is reported that enables the
efficient synthesis of previously inaccessible C4′-O-alkyl heptamethine cyanines. The key reaction involves N- to O- transposition with selective electrophile
incorporation on nitrogen. A representative fluorophore exhibits excellent
resistance to thiol nucleophiles, undergoes productive bioconjugation,
and can be used in near-IR fluorescence imaging applications.
Collapse
Affiliation(s)
- Roger R Nani
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health , Frederick, Maryland 21702, United States
| | | | | | | |
Collapse
|
39
|
Wang X, Li S, Shi Y, Chuan X, Li J, Zhong T, Zhang H, Dai W, He B, Zhang Q. The development of site-specific drug delivery nanocarriers based on receptor mediation. J Control Release 2014; 193:139-53. [DOI: 10.1016/j.jconrel.2014.05.028] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 05/13/2014] [Accepted: 05/17/2014] [Indexed: 01/28/2023]
|
40
|
Pearce AK, Rolfe BE, Russell PJ, Tse BWC, Whittaker AK, Fuchs AV, Thurecht KJ. Development of a polymer theranostic for prostate cancer. Polym Chem 2014. [DOI: 10.1039/c4py00999a] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
41
|
Minn I, Menezes ME, Sarkar S, Yarlagadda K, Das SK, Emdad L, Sarkar D, Fisher PB, Pomper MG. Molecular-genetic imaging of cancer. Adv Cancer Res 2014; 124:131-69. [PMID: 25287688 PMCID: PMC4339000 DOI: 10.1016/b978-0-12-411638-2.00004-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Molecular-genetic imaging of cancer using nonviral delivery systems has great potential for clinical application as a safe, efficient, noninvasive tool for visualization of various cellular processes including detection of cancer, and its attendant metastases. In recent years, significant effort has been expended in overcoming technical hurdles to enable clinical adoption of molecular-genetic imaging. This chapter will provide an introduction to the components of molecular-genetic imaging and recent advances on each component leading to safe, efficient clinical applications for detecting cancer. Combination with therapy, namely, generating molecular-genetic theranostic constructs, will provide further impetus for clinical translation of this promising technology.
Collapse
Affiliation(s)
- Il Minn
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Siddik Sarkar
- Department of Human and Molecular Genetics, Richmond, Virginia, USA
| | - Keerthi Yarlagadda
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Luni Emdad
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA.
| |
Collapse
|