1
|
Tomiyoshi K, Wilson LJ, Mourtada F, Mourtada JS, Namiki Y, Kamata W, Yang DJ, Inoue T. Optimization Processes of Clinical Chelation-Based Radiopharmaceuticals for Pathway-Directed Targeted Radionuclide Therapy in Oncology. Pharmaceutics 2024; 16:1458. [PMID: 39598580 PMCID: PMC11597032 DOI: 10.3390/pharmaceutics16111458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/09/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Targeted radionuclide therapy (TRT) for internal pathway-directed treatment is a game changer for precision medicine. TRT improves tumor control while minimizing damage to healthy tissue and extends the survival for patients with cancer. The application of theranostic-paired TRT along with cellular phenotype and genotype correlative analysis has the potential for malignant disease management. Chelation chemistry is essential for the development of theranostic-paired radiopharmaceuticals for TRT. Among image-guided TRT, 68Ga and 99mTc are the current standards for diagnostic radionuclides, while 177Lu and 225Ac have shown great promise for β- and α-TRT, respectively. Their long half-lives, potent radiobiology, favorable decay schemes, and ability to form stable chelation conjugates make them ideal for both manufacturing and clinical use. The current challenges include optimizing radionuclide production processes, coordinating chelation chemistry stability of theranostic-paired isotopes to reduce free daughters [this pertains to 225Ac daughters 221Fr and 213Bi]-induced tissue toxicity, and improving the modeling of micro dosimetry to refine dose-response evaluation. The empirical approach to TRT delivery is based on standard radionuclide administered activity levels, although clinical trials have revealed inconsistent outcomes and normal-tissue toxicities despite equivalent administered activities. This review presents the latest optimization methods for chelation-based theranostic radiopharmaceuticals, advancements in micro-dosimetry, and SPECT/CT technologies for quantifying whole-body uptake and monitoring therapeutic response as well as cytogenetic correlative analyses.
Collapse
Affiliation(s)
- Katsumi Tomiyoshi
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan
| | - Lydia J. Wilson
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA; (L.J.W.); (F.M.)
| | - Firas Mourtada
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA; (L.J.W.); (F.M.)
| | | | - Yuta Namiki
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan; (Y.N.); (W.K.); (D.J.Y.)
| | - Wataru Kamata
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan; (Y.N.); (W.K.); (D.J.Y.)
| | - David J. Yang
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan; (Y.N.); (W.K.); (D.J.Y.)
| | - Tomio Inoue
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan; (Y.N.); (W.K.); (D.J.Y.)
| |
Collapse
|
2
|
Wenker STM, van Lith SAM, Tamborino G, Konijnenberg MW, Bussink J, Heskamp S. The potential of targeted radionuclide therapy to treat hypoxic tumor cells. Nucl Med Biol 2024; 140-141:108971. [PMID: 39579561 DOI: 10.1016/j.nucmedbio.2024.108971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/25/2024]
Abstract
Tumor hypoxia contributes to cancer progression and therapy resistance. Several strategies have been investigated to relieve tumor hypoxia, of which some were successful. However, their clinical application remains challenging and therefore they are not used in daily clinical practice. Here, we review the potential of targeted radionuclide therapy (TRT) to eradicate hypoxic cancer cells. We present an overview of the published TRT strategies using β--particles, α-particles, and Auger electrons. Altogether, we conclude that α-particle emitting radionuclides are most promising since they can cause DNA double strand breaks independent of oxygen levels. Future directions for research are addressed, including more adequate in vitro and in vivo models to proof the potential of TRT to eliminate hypoxic cancer cells. Furthermore, dosimetry and radiobiology are identified as key to better understand the mechanism of action and dose-response relationships in hypoxic tumor areas. Finally, we can conclude that in order to achieve long-term anti-tumor efficacy, TRT combination treatment strategies may be necessary.
Collapse
Affiliation(s)
- S T M Wenker
- Department of Medical Imaging, Nuclear Medicine, Radboudumc, Nijmegen, the Netherlands; Department of Radiation Oncology, Radiotherapy & Oncoimmunology laboratory, Radboudumc, Nijmegen, the Netherlands
| | - S A M van Lith
- Department of Medical Imaging, Nuclear Medicine, Radboudumc, Nijmegen, the Netherlands
| | - G Tamborino
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - M W Konijnenberg
- Department of Medical Imaging, Nuclear Medicine, Radboudumc, Nijmegen, the Netherlands; Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - J Bussink
- Department of Radiation Oncology, Radiotherapy & Oncoimmunology laboratory, Radboudumc, Nijmegen, the Netherlands
| | - S Heskamp
- Department of Medical Imaging, Nuclear Medicine, Radboudumc, Nijmegen, the Netherlands.
| |
Collapse
|
3
|
Angelocci LV, Sgrignoli SS, de Souza CD, Antunes PCG, Rostelato MECM, Zeituni CA. In silicodosimetry for a prostate cancer treatment using 198Au nanoparticles. Biomed Phys Eng Express 2024; 11:015002. [PMID: 39447593 DOI: 10.1088/2057-1976/ad8acc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/24/2024] [Indexed: 10/26/2024]
Abstract
Objective. To estimate dose rates delivered by using radioactive198Au nanoparticles for prostate cancer nanobrachytherapy, identifying contribution by photons and electrons emmited from the source.Approach. Utilizingin silicomodels, two different anatomical representations were compared: a mathematical model and a unstructured mesh model based on the International Commission on Radiological Protection (ICRP) Publication 145 phantom. Dose rates by activity were calculated to the tumor and nearby healthy tissues, including healthy prostate tissue, urinary bladder wall and rectum, using Monte Carlo code MCNP6.2.Main results. Results indicate that both models provide dose rate estimates within the same order of magnitude, with the mathematical model overestimating doses to the prostate and bladder by approximately 20% compared to the unstructured mesh model. The discrepancies for the tumor and rectum were below 4%. Photons emmited from the source were defined as the primary contributors to dose to other organs, while 97.9% of the dose to the tumor was due to electrons emmited from the source.Significance. Our findings emphasize the importance of model selection in dosimetry, particularly the advantages of using realistic anatomical phantoms for accurate dose calculations. The study demonstrates the feasibility and effectiveness of198Au nanoparticles in achieving high dose concentrations in tumor regions while minimizing exposure to surrounding healthy tissues. Beta emissions were found to be predominantly responsible for tumor dose delivery, reinforcing the potential of198Au nanoparticles in localized radiation therapy. We advocate for using realistic body phantoms in further research to enhance reliability in dosimetry for nanobrachytherapy, as the field still lacks dedicated protocols.
Collapse
Affiliation(s)
- Lucas Verdi Angelocci
- Centro de Tecnologia das Radiações, Instituto de Pesquisas Energéticas e Nucleareas, São Paulo, SP, Brazil
| | | | - Carla Daruich de Souza
- Centro de Tecnologia das Radiações, Instituto de Pesquisas Energéticas e Nucleareas, São Paulo, SP, Brazil
- Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | | - Carlos Alberto Zeituni
- Centro de Tecnologia das Radiações, Instituto de Pesquisas Energéticas e Nucleareas, São Paulo, SP, Brazil
| |
Collapse
|
4
|
Luna Mass AC, Runge R, Wetzig K, Huebinger L, Brogsitter C, Kotzerke J. Enhancing Effects of Olaparib by Alpha- and Beta-Emitting Radionuclides, X-Rays, and Ultraviolet A Light in Combination with Ortho-IodoHoechst in a Prostate Cancer Cell Model. Pharmaceuticals (Basel) 2024; 17:1450. [PMID: 39598362 PMCID: PMC11597822 DOI: 10.3390/ph17111450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Background: New therapeutic strategies for metastatic castration-resistant prostate cancer (mCRPC) have been developed in the past to achieve the best response rates. Most recently, the use of combination therapies has been explored to optimize patient outcomes. Poly(ADP-ribose) polymerase inhibitors (PARPi) may help to treat mCRPC more effectively. Objectives: This study aimed to determine whether the combination of a PARPi with different radiation qualities results in different levels of radiosensitization of PC-3 cells. Methods: The radiosensitizing potential of Olaparib in combination with 177Lu, 223Ra, X-rays and photodynamic therapy (PDT) using the UVA light-activated photosensitizer ortho-iodoHoechst33258 (oIH) was evaluated by determining the clonogenic survival, DNA damage and cell cycle analysis. Results: Here, we show that this combination strategy differentially sensitized PC-3 cells to different radiation qualities. The combination of 177Lu with Olaparib increased the numbers of persistent double-strand breaks (DSBs) by a factor of 3.3 and cell death in PC-3 cells. Overall, the β-emitter 177Lu indicated a higher radiosensitization efficacy compared to 223Ra, with X-rays corresponding to dose modification factors (DMF) of 1.77, 1.17 and 1.16 respectively. Even in the case of the α-emitter 223Ra, the effects were much less pronounced than for 177Lu. PARPi also showed a slight potentiation of the cytotoxic effects both in co-treatment with X-rays and with PDT. Conclusions: The results of our study indicate a potential role for Olaparib in further optimizing the PSMA radioligand therapy (PRLT) outcomes. However, further evaluation of the combination of PARPi with PRLT is needed to gain more insights into improving the benefit to patients suffering from mCRPC.
Collapse
Affiliation(s)
- Andrea C. Luna Mass
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, D-01307 Dresden, Germany; (R.R.); (K.W.); (L.H.); (C.B.); (J.K.)
| | | | | | | | | | | |
Collapse
|
5
|
Andrew J, Ezra-Manicum AL, Witika BA. Developments in radionanotheranostic strategies for precision diagnosis and treatment of prostate cancer. EJNMMI Radiopharm Chem 2024; 9:62. [PMID: 39180599 PMCID: PMC11344754 DOI: 10.1186/s41181-024-00295-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/13/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Prostate Cancer (PCa) is the second most diagnosed urological cancer among men worldwide. Conventional methods used for diagnosis of PCa have several pitfalls which include lack of sensitivity and specificity. On the other hand, traditional treatment of PCa poses challenges such as long-term side effects and the development of multidrug resistance (MDR). MAIN BODY Hence, there is a need for novel PCa agents with the potential to lessen the burden of these adverse effects on patients. Nanotechnology has emerged as a promising approach to support both early diagnosis and effective treatment of tumours by ensuring precise delivery of the drug to the targeted site of the disease. Most cancer-related biological processes occur on the nanoscale hence application of nanotechnology has been greatly appreciated and implemented in the management and therapeutics of cancer. Nuclear medicine plays a significant role in the non-invasive diagnosis and treatment of PCa using appropriate radiopharmaceuticals. This review aims to explore the different radiolabelled nanomaterials to enhance the specific delivery of imaging and therapeutic agents to cancer cells. Thereafter, the review appraises the advantages and disadvantages of these modalities and then discusses and outlines the benefits of radiolabelled nanomaterials in targeting cancerous prostatic tumours. Moreover, the nanoradiotheranostic approaches currently developed for PCa are discussed and finally the prospects of combining radiopharmaceuticals with nanotechnology in improving PCa outcomes will be highlighted. CONCLUSION Nanomaterials have great potential, but safety and biocompatibility issues remain. Notwithstanding, the combination of nanomaterials with radiotherapeutics may improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Jubilee Andrew
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, 0208, South Africa
| | - Amanda-Lee Ezra-Manicum
- Department of Chemistry, Faculty of Science, Tshwane University of Technology (Arcadia Campus), Pretoria, South Africa
| | - Bwalya Angel Witika
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, 0208, South Africa.
| |
Collapse
|
6
|
Vosoughi S, Salek N, Zolghadri S, Aghamiri SMR, Delavari M. Optimized Production of 188Re-HYNIC-Bombesin: New Therapeutic Agent for GRPR Targeting. Nucl Med Mol Imaging 2024; 58:300-309. [PMID: 39036461 PMCID: PMC11255143 DOI: 10.1007/s13139-024-00866-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/22/2024] [Accepted: 05/14/2024] [Indexed: 07/23/2024] Open
Abstract
Purpose One of the most interesting methods to deliver therapeutic doses of ionizing radiation to tumor sites is radiolabeled compounds. Bombesin peptide binds to gastrin-releasing peptide receptors (GRPRs) with great affinity. Through its appropriate physical characteristics and accessibility as the 188W/188Re generator, 188Re can be effectively used to develop a therapeutic radio complex. In this study, 188Re-HYNIC-BBN was prepared under optimal conditions. Methods Optimization of the effective parameters on 188Re-HYNIC-BBN radio-labeling yield like ligand concentration, pH, reaction time, and temperature were performed. The final product's radiochemical purity was measured by RTLC and HPLC. The stability of the radio-complex was checked in PBS buffer (4 °C) and human blood serum (37 °C). The partition coefficient of the final radio-complex was studied using standard procedure. Finally, the biodistribution of 188Re-HYNIC-BBN and free 188Re in different organs of the rats were compared in various intervals. Results The final product was prepared with a specific activity of 7.11 TBq/mmol and radiochemical purity > 95% at the optimized conditions (pH = 4-5, reaction time = 45 min, temp = 95℃). This radio-complex was found to be stable in PBS and blood serum over 24 h. LogPo/w was - 1.78, showing the high hydrophilic nature of the radio-complex. The biodistribution of 188Re-HYNIC-BBN demonstrated the fast clearance of the radio-peptide from the blood circulation. The most portion of the radioactivity was excreted from the body via the urinary tract and the remaining activity was accumulated in GRPR-expressing organs. Conclusion The special characteristics of the complex introduce 188Re-HYNIC-BBN as a new therapeutic agent for targeting GRPRs, however, more biological data is still needed.
Collapse
Affiliation(s)
- Sara Vosoughi
- Radiation Application Research School, Nuclear Science and Technology Research Institute (NSTRI), Box 14395‑836, North Kargar Street, Tehran, Iran
| | - Nafise Salek
- Nuclear Fuel Research School, Nuclear Science and Technology Research Institute (NSTRI), Box 14395‑836, North Kargar Street, Tehran, Iran
| | - Samaneh Zolghadri
- Radiation Application Research School, Nuclear Science and Technology Research Institute (NSTRI), Box 14395‑836, North Kargar Street, Tehran, Iran
| | | | - Milad Delavari
- Radiation Application Research School, Nuclear Science and Technology Research Institute (NSTRI), Box 14395‑836, North Kargar Street, Tehran, Iran
| |
Collapse
|
7
|
Schatz CA, Zitzmann-Kolbe S, Moen I, Klotz M, Nair S, Stargard S, Bjerke RM, Wickstrøm Biseth K, Feng YZ, Indrevoll B, Cruciani V, Karlsson J, Haendler B, Nielsen CH, Alfsen MZ, Hammer S, Hennekes H, Cuthbertson A, Hagemann UB, Larsen Å. Preclinical Efficacy of a PSMA-Targeted Actinium-225 Conjugate (225Ac-Macropa-Pelgifatamab): A Targeted Alpha Therapy for Prostate Cancer. Clin Cancer Res 2024; 30:2531-2544. [PMID: 38593212 DOI: 10.1158/1078-0432.ccr-23-3746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/07/2024] [Accepted: 04/03/2024] [Indexed: 04/11/2024]
Abstract
PURPOSE Initially, prostate cancer responds to hormone therapy, but eventually resistance develops. Beta emitter-based prostate-specific membrane antigen (PSMA)-targeted radionuclide therapy is approved for the treatment of metastatic castration-resistant prostate cancer. Here we introduce a targeted alpha therapy (TAT) consisting of the PSMA antibody pelgifatamab covalently linked to a macropa chelator and labeled with actinium-225 and compare its efficacy and tolerability with other TATs. EXPERIMENTAL DESIGN The in vitro characteristics and in vivo biodistribution, antitumor efficacy, and tolerability of 225Ac-macropa-pelgifatamab (225Ac-pelgi) and other TATs were investigated in cell line- and patient-derived prostate cancer xenograft models. The antitumor efficacy of 225Ac-pelgi was also investigated in combination with the androgen receptor inhibitor darolutamide. RESULTS Actinium-225-labeling of 225Ac-pelgi was efficient already at room temperature. Potent in vitro cytotoxicity was seen in PSMA-expressing (LNCaP, MDA-PCa-2b, and C4-2) but not in PSMA-negative (PC-3 and DU-145) cell lines. High tumor accumulation was seen for both 225Ac-pelgi and 225Ac-DOTA-pelgi in the MDA-PCa-2b xenograft model. In the C4-2 xenograft model, 225Ac-pelgi showed enhanced antitumor efficacy with a T/Cvolume (treatment/control) ratio of 0.10 compared with 225Ac-DOTA-pelgi, 225Ac-DOTA-J591, and 227Th-HOPO-pelgifatamab (227Th-pelgi; all at 300 kBq/kg) with T/Cvolume ratios of 0.37, 0.39, and 0.33, respectively. 225Ac-pelgi was less myelosuppressive than 227Th-pelgi. 225Ac-pelgi showed dose-dependent treatment efficacy in the patient-derived KuCaP-1 model and strong combination potential with darolutamide in both cell line- (22Rv1) and patient-derived (ST1273) xenograft models. CONCLUSIONS These results provide a strong rationale to investigate 225Ac-pelgi in patients with prostate cancer. A clinical phase I study has been initiated (NCT06052306).
Collapse
|
8
|
Chiu CH, Yang DJ, Liou YC, Chang WC, Yu TH, Chung MC, Lee YC, Chen IJ, Wang PY, Lin CP, Tsay HJ, Yeh SHH. Assessment of DNA/RNA Deregulation in Cancer Using 99mTc-Labeled Thiopurine. Cancer Biother Radiopharm 2024; 39:358-372. [PMID: 38457659 DOI: 10.1089/cbr.2023.0148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2024] Open
Affiliation(s)
- Chuang-Hsin Chiu
- Department of Nuclear Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - David J Yang
- Seecure/Taiwan Hopax Chemicals MFG Company Ltd., Kaohsiung, Taiwan
| | - Yi-Chen Liou
- Institute of Neuroscience, National Chaio Tung University, Taipei, Taiwan
| | - Wei-Chung Chang
- Seecure/Taiwan Hopax Chemicals MFG Company Ltd., Kaohsiung, Taiwan
| | - Tsung-Hsun Yu
- Brain Research Center, National Chaio Tung University, Taipei, Taiwan
- Brain Research Center, School of Medicine, National Defense Medical Center Taipei, Taiwan
| | - Min-Ching Chung
- Seecure/Taiwan Hopax Chemicals MFG Company Ltd., Kaohsiung, Taiwan
| | - Yen-Chun Lee
- Seecure/Taiwan Hopax Chemicals MFG Company Ltd., Kaohsiung, Taiwan
| | - Ing-Jou Chen
- Department of Nuclear Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Pao-Yeh Wang
- Brain Research Center, School of Medicine, National Defense Medical Center Taipei, Taiwan
| | - Ching-Po Lin
- Institute of Neuroscience, National Chaio Tung University, Taipei, Taiwan
| | - Hey-Jen Tsay
- Institute of Neuroscience, National Chaio Tung University, Taipei, Taiwan
| | - Skye Hsin-Hsien Yeh
- Brain Research Center, National Chaio Tung University, Taipei, Taiwan
- Brain Research Center, School of Medicine, National Defense Medical Center Taipei, Taiwan
| |
Collapse
|
9
|
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
|
10
|
Xiao D, Han P, Jiang Y, Duan X, Ruan Q, Zhang Z, Chen X, Zhang J. Preparation, Biological Evaluation, and First-in-Human Single-Photon Emission Computed Tomography (SPECT) Study of 99mTc-Labeled Prostate-Specific Membrane Antigen (PSMA)-Targeted Radiotracers Containing Triazole with Reduced Kidneys Accumulation. ACS Pharmacol Transl Sci 2024; 7:1335-1347. [PMID: 38751628 PMCID: PMC11092200 DOI: 10.1021/acsptsci.4c00012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/16/2024] [Accepted: 03/27/2024] [Indexed: 05/18/2024]
Abstract
Prostate-specific membrane antigen (PSMA), a well-established biological marker for prostate cancer (PCa) imaging and therapy, is overexpressed on the surface of prostate cancer lesions. In this study, a triazole ring was introduced into the linker by click chemistry to generate a HYNIC-derived ligand (T), which exhibited good PSMA affinity (Ki = 2.23 nM). Eight stable 99mTc-labeled complexes, [99mTc]Tc-T-Mn (n = 1-8), with hydrophilic properties were synthesized by incorporating different coligands at high radiochemical yields and purities without purification. The radioligands were concentrated in the kidneys of healthy Kunming male mice and were significantly blocked by the PSMA inhibitor ZJ-43. The uptake of the optimized complex [99mTc]Tc-T-M2 was correlated with PSMA, and it had good PSMA affinity (Kd = 5.42 nM). [99mTc]Tc-T-M2 accumulated on LNCaP (PSMA++) tumors and was significantly blocked by ZJ-43 at 2 h p.i., indicating high PSMA specificity. Relatively suitable kidney uptake was beneficial for reducing kidneys exposure in patients. SPECT/CT imaging of [99mTc]Tc-T-M2 in LNCaP (PSMA++) or 22Rv1 (PSMA+) tumor-bearing mice revealed high tumor uptake, low background uptake (especially low kidney uptake (49.06 ± 9.20 %ID/g) at 2 h p.i.), and obvious inhibition by ZJ-43, whereas PC-3 (PSMA-) tumors were undetectable. A freeze-dried [99mTc]Tc-T-M2 kit was successfully developed (T-M2 kit). Preliminary clinical trials showed that [99mTc]Tc-T-M2 clearly identified small prostate cancer lesions and has potential for clinical application.
Collapse
Affiliation(s)
- Di Xiao
- 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
| | - Peiwen Han
- 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
| | - 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
| | - Xiaojiang Duan
- Department
of Nuclear Medicine, Peking University First
Hospital, Beijing 100034, 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
| | - Zhanbin 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
| | - Xiaoliang Chen
- Department
of Nuclear Medicine, Chongqing University
Cancer Hospital, Chongqing 400030, P.R. China
- Hubei
Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji
Medical College, Huazhong University of
Science and Technology, Wuhan 430022, 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
|
11
|
Taş H, Bakos G, Bauder-Wüst U, Schäfer M, Remde Y, Roscher M, Benešová-Schäfer M. Human ABC and SLC Transporters: The Culprit Responsible for Unspecific PSMA-617 Uptake? Pharmaceuticals (Basel) 2024; 17:513. [PMID: 38675472 PMCID: PMC11053447 DOI: 10.3390/ph17040513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
[177Lu]Lu-PSMA-617 has recently been successfully approved by the FDA, the MHRA, Health Canada and the EMA as Pluvicto®. However, salivary gland (SG) and kidney toxicities account for its main dose-limiting side-effects, while its corresponding uptake and retention mechanisms still remain elusive. Recently, the presence of different ATP-binding cassette (ABC) transporters, such as human breast cancer resistance proteins (BCRP), multidrug resistance proteins (MDR1), multidrug-resistance-related proteins (MRP1, MRP4) and solute cassette (SLC) transporters, such as multidrug and toxin extrusion proteins (MATE1, MATE2-K), organic anion transporters (OAT1, OAT2v1, OAT3, OAT4) and peptide transporters (PEPT2), has been verified at different abundances in human SGs and kidneys. Therefore, our aim was to assess whether [177Lu]Lu-PSMA-617 and [225Ac]Ac-PSMA-617 are substrates of these ABC and SLC transporters. For in vitro studies, the novel isotopologue ([α,β-3H]Nal)Lu-PSMA-617 was used in cell lines or vesicles expressing the aforementioned human ABC and SLC transporters for inhibition and uptake studies, respectively. The corresponding probe substrates and reference inhibitors were used as controls. Our results indicate that [177Lu]Lu-PSMA-617 and [225Ac]Ac-PSMA-617 are neither inhibitors nor substrates of the examined transporters. Therefore, our results show that human ABC and SLC transporters play no central role in the uptake and retention of [177Lu]Lu-PSMA-617 and [225Ac]Ac-PSMA-617 in the SGs and kidneys nor in the observed toxicities.
Collapse
Affiliation(s)
- Harun Taş
- German Cancer Research Center (DKFZ), Research Group Molecular Biology of Systemic Radiotherapy, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (H.T.); (G.B.); (U.B.-W.)
| | - Gábor Bakos
- German Cancer Research Center (DKFZ), Research Group Molecular Biology of Systemic Radiotherapy, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (H.T.); (G.B.); (U.B.-W.)
| | - Ulrike Bauder-Wüst
- German Cancer Research Center (DKFZ), Research Group Molecular Biology of Systemic Radiotherapy, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (H.T.); (G.B.); (U.B.-W.)
| | - Martin Schäfer
- German Cancer Research Center (DKFZ), Service Unit for Radiopharmaceuticals and Preclinical Trials, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (M.S.); (Y.R.); (M.R.)
| | - Yvonne Remde
- German Cancer Research Center (DKFZ), Service Unit for Radiopharmaceuticals and Preclinical Trials, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (M.S.); (Y.R.); (M.R.)
| | - Mareike Roscher
- German Cancer Research Center (DKFZ), Service Unit for Radiopharmaceuticals and Preclinical Trials, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (M.S.); (Y.R.); (M.R.)
| | - Martina Benešová-Schäfer
- German Cancer Research Center (DKFZ), Research Group Molecular Biology of Systemic Radiotherapy, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (H.T.); (G.B.); (U.B.-W.)
| |
Collapse
|
12
|
Shea AG, Idrissou MB, Torres AI, Chen T, Hernandez R, Morris ZS, Sodji QH. Immunological effects of radiopharmaceutical therapy. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 4:1331364. [PMID: 39355211 PMCID: PMC11440989 DOI: 10.3389/fnume.2024.1331364] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/14/2024] [Indexed: 10/03/2024]
Abstract
Radiation therapy (RT) is a pillar of cancer therapy used by more than half of all cancer patients. Clinically, RT is mostly delivered as external beam radiation therapy (EBRT). However, the scope of EBRT is limited in the metastatic setting, where all sites of disease need to be irradiated. Such a limitation is attributed to radiation-induced toxicities, for example on bone marrow and hematologic toxicities, resulting from a large EBRT field. Radiopharmaceutical therapy (RPT) has emerged as an alternative to EBRT for the irradiation of all sites of metastatic disease. While RPT can reduce tumor burden, it can also impact the immune system and anti-tumor immunity. Understanding these effects is crucial for predicting and managing treatment-related hematological toxicities and optimizing their integration with other therapeutic modalities, such as immunotherapies. Here, we review the immunomodulatory effects of α- and β-particle emitter-based RPT on various immune cell lines, such as CD8+ and CD4+ T cells, natural killer (NK) cells, and regulatory T (Treg) cells. We briefly discuss Auger electron-emitter (AEE)-based RPT, and finally, we highlight the combination of RPT with immune checkpoint inhibitors, which may offer potential therapeutic synergies for patients with metastatic cancers.
Collapse
Affiliation(s)
- Amanda G. Shea
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Malick Bio Idrissou
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Ana Isabel Torres
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Tessa Chen
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Reiner Hernandez
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Zachary S. Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Quaovi H. Sodji
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
13
|
Ghosh S, Lee SJ, Hsu JC, Chakraborty S, Chakravarty R, Cai W. Cancer Brachytherapy at the Nanoscale: An Emerging Paradigm. CHEMICAL & BIOMEDICAL IMAGING 2024; 2:4-26. [PMID: 38274040 PMCID: PMC10806911 DOI: 10.1021/cbmi.3c00092] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/09/2023] [Accepted: 11/01/2023] [Indexed: 01/27/2024]
Abstract
Brachytherapy is an established treatment modality that has been globally utilized for the therapy of malignant solid tumors. However, classic therapeutic sealed sources used in brachytherapy must be surgically implanted directly into the tumor site and removed after the requisite period of treatment. In order to avoid the trauma involved in the surgical procedures and prevent undesirable radioactive distribution at the cancerous site, well-dispersed radiolabeled nanomaterials are now being explored for brachytherapy applications. This emerging field has been coined "nanoscale brachytherapy". Despite present-day advancements, an ongoing challenge is obtaining an advanced, functional nanomaterial that concurrently incorporates features of high radiolabeling yield, short labeling time, good radiolabeling stability, and long tumor retention time without leakage of radioactivity to the nontargeted organs. Further, attachment of suitable targeting ligands to the nanoplatforms would widen the nanoscale brachytherapy approach to tumors expressing various phenotypes. Molecular imaging using radiolabeled nanoplatforms enables noninvasive visualization of cellular functions and biological processes in vivo. In vivo imaging also aids in visualizing the localization and retention of the radiolabeled nanoplatforms at the tumor site for the requisite time period to render safe and effective therapy. Herein, we review the advancements over the last several years in the synthesis and use of functionalized radiolabeled nanoplatforms as a noninvasive substitute to standard brachytherapy sources. The limitations of present-day brachytherapy sealed sources are analyzed, while highlighting the advantages of using radiolabeled nanoparticles (NPs) for this purpose. The recent progress in the development of different radiolabeling methods, delivery techniques and nanoparticle internalization mechanisms are discussed. The preclinical studies performed to date are summarized with an emphasis on the current challenges toward the future translation of nanoscale brachytherapy in routine clinical practices.
Collapse
Affiliation(s)
- Sanchita Ghosh
- Radiopharmaceuticals
Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Homi
Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Sophia J. Lee
- Departments
of Radiology and Medical Physics, University
of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jessica C. Hsu
- Departments
of Radiology and Medical Physics, University
of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Sudipta Chakraborty
- Radiopharmaceuticals
Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Homi
Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Rubel Chakravarty
- Radiopharmaceuticals
Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Homi
Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Weibo Cai
- Departments
of Radiology and Medical Physics, University
of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| |
Collapse
|
14
|
Musket A, Davern S, Elam BM, Musich PR, Moorman JP, Jiang Y. The application of radionuclide therapy for breast cancer. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 3:1323514. [PMID: 39355029 PMCID: PMC11440853 DOI: 10.3389/fnume.2023.1323514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/27/2023] [Indexed: 10/03/2024]
Abstract
Radionuclide-mediated diagnosis and therapy have emerged as effective and low-risk approaches to treating breast cancer. Compared to traditional anatomic imaging techniques, diagnostic radionuclide-based molecular imaging systems exhibit much greater sensitivity and ability to precisely illustrate the biodistribution and metabolic processes from a functional perspective in breast cancer; this transitions diagnosis from an invasive visualization to a noninvasive visualization, potentially ensuring earlier diagnosis and on-time treatment. Radionuclide therapy is a newly developed modality for the treatment of breast cancer in which radionuclides are delivered to tumors and/or tumor-associated targets either directly or using delivery vehicles. Radionuclide therapy has been proven to be eminently effective and to exhibit low toxicity when eliminating both primary tumors and metastases and even undetected tumors. In addition, the specific interaction between the surface modules of the delivery vehicles and the targets on the surface of tumor cells enables radionuclide targeting therapy, and this represents an exceptional potential for this treatment in breast cancer. This article reviews the development of radionuclide molecular imaging techniques that are currently employed for early breast cancer diagnosis and both the progress and challenges of radionuclide therapy employed in breast cancer treatment.
Collapse
Affiliation(s)
- Anna Musket
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Sandra Davern
- Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Brianna M Elam
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Philip R Musich
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Jonathan P Moorman
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Yong Jiang
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| |
Collapse
|
15
|
Zhang H, Xiao L, Xie H, Li L. Hotspots and frontiers in PSMA research for prostate cancer: a bibliometric and visualization analysis over the past 20 years. Eur J Med Res 2023; 28:610. [PMID: 38115121 PMCID: PMC10731714 DOI: 10.1186/s40001-023-01590-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/12/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Prostate-specific membrane antigen (PSMA)-targeted imaging and therapy have significantly changed the management of patients with prostate cancer (PCa) at different disease stages. This advancement has attracted the attention of scholars, leading to a prolific output of scholarly publications. This study comprehensively outlines the knowledge framework associated with PSMA-based diagnosis and treatment of PCa through the application of bibliometric analysis, and discusses the potential research trends and foci. METHODS Articles and reviews related to PSMA for prostate cancer from 2003 to 2022 were retrieved from Web of Science Core Collection. VOSviewer, Citespace, and R-bibliometrix were primarily employed to execute and visually represent co-authorship, co-citation, and co-occurrence analysis of countries, institutions, authors, references and keywords in this field. RESULTS A total of 3830 papers were included. The papers on the field of PSMA-based PCa therapy and imaging had been continuously increased since 2003, but the rate has slowed from 2020. The United States made the largest contribution in this field, in terms of publications 997 (26.03%), H-index (110) and total citations (53,167 times). We identified the most productive institution were Technical University of Munich, and Australian institutions had become very active in recent years. Journal of Nuclear Medicine was the most prominent journal in this field. Professors Matthias Eiber and Martin G Pomper made great achievements, while Ali Afshar-Oromieh was the most co-cited author. According to the result of keywords and topics analysis, "ga-68 labeled psma ligand", "radiation dosimetry" and "HBED-CC" were major research areas in the near future, while "Extended pelvic lymph node dissection" was considered to be the future research foci. CONCLUSIONS The field of psma-based PCa therapy and imaging is in the stage of vigorous development and has a bright prospect. The United States and Germany have achieved outstanding results in this area, while Australia has recently developed rapidly. It is foreseeable that more research foci will be lied in the early detection of pelvic lymph nodes and the multimodal imaging-guided surgery.
Collapse
Affiliation(s)
- Hanfei Zhang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, 610041, China
| | - Liu Xiao
- Department of Nuclear Medicine, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, 610041, China
| | - Hangyu Xie
- Department of Nuclear Medicine, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, 610041, China
| | - Lin Li
- Department of Nuclear Medicine, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, 610041, China.
| |
Collapse
|
16
|
Mourtada F, Tomiyoshi K, Sims-Mourtada J, Mukai-Sasaki Y, Yagihashi T, Namiki Y, Murai T, Yang DJ, Inoue T. Actinium-225 Targeted Agents: Where Are We Now? Brachytherapy 2023; 22:697-708. [PMID: 37690972 PMCID: PMC10840862 DOI: 10.1016/j.brachy.2023.06.228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 06/26/2023] [Indexed: 09/12/2023]
Abstract
α-particle targeted radionuclide therapy has shown promise for optimal cancer management, an exciting new era for brachytherapy. Alpha-emitting nuclides can have significant advantages over gamma- and beta-emitters due to their high linear energy transfer (LET). While their limited path length results in more specific tumor 0kill with less damage to surrounding normal tissues, their high LET can produce substantially more lethal double strand DNA breaks per radiation track than beta particles. Over the last decade, the physical and chemical attributes of Actinium-225 (225Ac) including its half-life, decay schemes, path length, and straightforward chelation ability has peaked interest for brachytherapy agent development. However, this has been met with challenges including source availability, accurate modeling for standardized dosimetry for brachytherapy treatment planning, and laboratory space allocation in the hospital setting for on-demand radiopharmaceuticals production. Current evidence suggests that a simple empirical approach based on 225Ac administered radioactivity may lead to inconsistent outcomes and toxicity. In this review article, we highlight the recent advances in 225Ac source production, dosimetry modeling, and current clinical studies.
Collapse
Affiliation(s)
- Firas Mourtada
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA.
| | - Katsumi Tomiyoshi
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | | | - Yuki Mukai-Sasaki
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan; Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Takayuki Yagihashi
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Yuta Namiki
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Taro Murai
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - David J Yang
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Tomio Inoue
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan
| |
Collapse
|
17
|
Ritawidya R, Wongso H, Effendi N, Pujiyanto A, Lestari W, Setiawan H, Humani TS. Lutetium-177-Labeled Prostate-Specific Membrane Antigen-617 for Molecular Imaging and Targeted Radioligand Therapy of Prostate Cancer. Adv Pharm Bull 2023; 13:701-711. [PMID: 38022814 PMCID: PMC10676551 DOI: 10.34172/apb.2023.079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 02/04/2023] [Accepted: 04/24/2023] [Indexed: 12/01/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA) represents a promising target for PSMA-overexpressing diseases, especially prostate cancer-a common type of cancer among men worldwide. In response to the challenges in tackling prostate cancers, several promising PSMA inhibitors from a variety of molecular scaffolds (e.g., phosphorous-, thiol-, and urea-based molecules) have been developed. In addition, PSMA inhibitors bearing macrocyclic chelators have attracted interest due to their favorable pharmacokinetic properties. Recently, conjugating a small PSMA molecule inhibitor-bearing 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) chelator, as exemplified by [177Lu]Lu-PSMA-617 could serve as a molecular imaging probe and targeted radioligand therapy (TRT) of metastatic castration resistant prostate cancer (mCRPC). Hence, studies related to mCRPC have drawn global attention. In this review, the recent development of PSMA ligand-617-labeled with 177Lu for the management of mCRPC is presented. Its molecular mechanism of action, safety, efficacy, and future direction are also described.
Collapse
Affiliation(s)
- Rien Ritawidya
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, National Research and Innovation Agency (BRIN), Kawasan Puspiptek, Setu, Tangerang Selatan, 15314 Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Raya Bandung-Sumedang KM 21, Sumedang, 45363, Indonesia
| | - Hendris Wongso
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, National Research and Innovation Agency (BRIN), Kawasan Puspiptek, Setu, Tangerang Selatan, 15314 Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Raya Bandung-Sumedang KM 21, Sumedang, 45363, Indonesia
| | - Nurmaya Effendi
- Faculty of Pharmacy, University of Muslim Indonesia, Kampus II UMI, Jl. Urip Sumoharjo No.225, Panaikang, Panakkukang, Kota, Makassar, Sulawesi Selatan 90231
| | - Anung Pujiyanto
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, National Research and Innovation Agency (BRIN), Kawasan Puspiptek, Setu, Tangerang Selatan, 15314 Indonesia
| | - Wening Lestari
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, National Research and Innovation Agency (BRIN), Kawasan Puspiptek, Setu, Tangerang Selatan, 15314 Indonesia
| | - Herlan Setiawan
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, National Research and Innovation Agency (BRIN), Kawasan Puspiptek, Setu, Tangerang Selatan, 15314 Indonesia
| | - Titis Sekar Humani
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, National Research and Innovation Agency (BRIN), Kawasan Puspiptek, Setu, Tangerang Selatan, 15314 Indonesia
| |
Collapse
|
18
|
Baun C, Dam JH, Hildebrandt MG, Ewald JD, Kristensen BW, Gammelsrød VS, Olsen BB, Thisgaard H. Preclinical evaluation of [ 58mCo]Co-DOTA-PSMA-617 for Auger electron therapy of prostate cancer. Sci Rep 2023; 13:18837. [PMID: 37914790 PMCID: PMC10620164 DOI: 10.1038/s41598-023-43429-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/23/2023] [Indexed: 11/03/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA), highly expressed in prostate cancer, is a promising target for radionuclide therapy. Auger electron-emitting radionuclides are well suited for targeted radionuclide therapy if they can be delivered close to the DNA of the targeted cells. This preclinical study evaluated the theranostic pair [55/58mCo]Co-DOTA-PSMA-617 for PET imaging and Auger electron therapy of prostate cancer. [58mCo]Co-DOTA-PSMA-617 was successfully prepared with > 99% radiochemical yield and purity. In vitro, uptake and subcellular distribution assays in PSMA-positive prostate cancer cells showed PSMA-specific uptake with high cell-associated activity in the nucleus. Incubation with [58mCo]Co-DOTA-PSMA-617 reduced cell viability and clonogenic survival in a significant dose-dependent manner (p < 0.05). Biodistribution of xenografted mice showed high specific tumor uptake of the cobalt-labeled PSMA ligand for all time points with rapid clearance from normal tissues, which PET imaging confirmed. In vivo, therapy with [58mCo]Co-DOTA-PSMA-617 in tumor-bearing mice demonstrated significantly increased median survival for treated mice compared to control animals (p = 0.0014). In conclusion, [55/58mCo]Co-DOTA-PSMA-617 displayed excellent in vitro and in vivo properties, offering significant survival benefits in mice with no observed toxicities.
Collapse
Affiliation(s)
- Christina Baun
- Department of Nuclear Medicine, Odense University Hospital, Kløvervænget 47, 5000, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Center for Personalized Response Monitoring in Oncology (PREMIO), Odense University Hospital, Odense, Denmark
| | - Johan Hygum Dam
- Department of Nuclear Medicine, Odense University Hospital, Kløvervænget 47, 5000, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Malene Grubbe Hildebrandt
- Department of Nuclear Medicine, Odense University Hospital, Kløvervænget 47, 5000, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Center for Personalized Response Monitoring in Oncology (PREMIO), Odense University Hospital, Odense, Denmark
- Centre for Innovative Medical Technology, Odense University Hospital, Odense, Denmark
| | - Jesper Dupont Ewald
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Bjarne Winther Kristensen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Vigga Sand Gammelsrød
- Department of Nuclear Medicine, Odense University Hospital, Kløvervænget 47, 5000, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Birgitte Brinkmann Olsen
- Department of Nuclear Medicine, Odense University Hospital, Kløvervænget 47, 5000, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Surgical Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Helge Thisgaard
- Department of Nuclear Medicine, Odense University Hospital, Kløvervænget 47, 5000, Odense C, Denmark.
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
19
|
Navarro Ledesma S, Hamed-Hamed D, González-Muñoz A, Pruimboom L. Effectiveness of Treatments That Alter Metabolomics in Cancer Patients-A Systematic Review. Cancers (Basel) 2023; 15:4297. [PMID: 37686573 PMCID: PMC10486463 DOI: 10.3390/cancers15174297] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/07/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
INTRODUCTION Cancer is the leading cause of death worldwide, with the most frequent being breast cancer in women, prostate cancer in men and colon cancer in both sexes. The use of metabolomics to find new biomarkers can provide knowledge about possible interventions based on the presence of oncometabolites in different cancer types. OBJECTIVES The primary purpose of this review is to analyze the characteristic metabolome of three of the most frequent cancer types. We further want to identify the existence and success rate of metabolomics-based intervention in patients suffering from those cancer types. Our conclusions are based on the analysis of the methodological quality of the studies. METHODS We searched for studies that investigated the metabolomic characteristics in patients suffering from breast cancer, prostate cancer or colon cancer in clinical trials. The data were analyzed, as well as the effects of specific interventions based on identified metabolomics and one or more oncometabolites. The used databases were PubMed, Virtual Health Library, Web of Science, EBSCO and Cochrane Library. Only nine studies met the selection criteria. Study bias was analyzed using the Cochrane risk of bias tool. This systematic review protocol was registered at the International Prospective Register of Systematic Reviews (PROSPERO: CRD42023401474). RESULTS Only nine studies about clinical trials were included in this review and show a moderate quality of evidence. Metabolomics-based interventions related with disease outcome were conflictive with no or small changes in the metabolic characteristics of the different cancer types. CONCLUSIONS This systematic review shows some interesting results related with metabolomics-based interventions and their effects on changes in certain cancer oncometabolites. The small number of studies we identified which fulfilled our inclusion criteria in this systematic review does not allow us to draw definitive conclusions. Nevertheless, some results can be considered as promising although further research is needed. That research must focus not only on the presence of possible oncometabolites but also on possible metabolomics-based interventions and their influence on the outcome in patients suffering from breast cancer, prostate cancer or colon cancer.
Collapse
Affiliation(s)
- Santiago Navarro Ledesma
- Department of Physiotherapy, Faculty of Health Sciences, Campus of Melilla, University of Granada, Querol Street 5, 52004 Melilla, Spain; (D.H.-H.); (A.G.-M.)
- Department of Physiotherapy, University Chair in Clinical Psychoneuroimmunology, University of Granada and PNI Europe, 52004 Melilla, Spain;
| | - Dina Hamed-Hamed
- Department of Physiotherapy, Faculty of Health Sciences, Campus of Melilla, University of Granada, Querol Street 5, 52004 Melilla, Spain; (D.H.-H.); (A.G.-M.)
| | - Ana González-Muñoz
- Department of Physiotherapy, Faculty of Health Sciences, Campus of Melilla, University of Granada, Querol Street 5, 52004 Melilla, Spain; (D.H.-H.); (A.G.-M.)
| | - Leo Pruimboom
- Department of Physiotherapy, University Chair in Clinical Psychoneuroimmunology, University of Granada and PNI Europe, 52004 Melilla, Spain;
| |
Collapse
|
20
|
Thakur N, Quazi S, Naik B, Jha SK, Singh P. New insights into molecular signaling pathways and current advancements in prostate cancer diagnostics & therapeutics. Front Oncol 2023; 13:1193736. [PMID: 37664036 PMCID: PMC10469924 DOI: 10.3389/fonc.2023.1193736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 07/18/2023] [Indexed: 09/05/2023] Open
Abstract
Prostate adenocarcinoma accounts for more than 20% of deaths among males due to cancer. It is the fifth-leading cancer diagnosed in males across the globe. The mortality rate is quite high due to prostate cancer. Despite the fact that advancements in diagnostics and therapeutics have been made, there is a lack of effective drugs. Metabolic pathways are altered due to the triggering of androgen receptor (AR) signaling pathways, and elevated levels of dihydrotestosterone are produced due to defects in AR signaling that accelerate the growth of prostate cancer cells. Further, PI3K/AKT/mTOR pathways interact with AR signaling pathway and act as precursors to promote prostate cancer. Prostate cancer therapy has been classified into luminal A, luminal B, and basal subtypes. Therapeutic drugs inhibiting dihydrotestosterone and PI3K have shown to give promising results to combat prostate cancer. Many second-generation Androgen receptor signaling antagonists are given either as single agent or with the combination of other drugs. In order to develop a cure for metastasized prostate cancer cells, Androgen deprivation therapy (ADT) is applied by using surgical or chemical methods. In many cases, Prostatectomy or local radiotherapy are used to control metastasized prostate cancer. However, it has been observed that after 1.5 years to 2 years of Prostatectomy or castration, there is reoccurrence of prostate cancer and high incidence of castration resistant prostate cancer is seen in population undergone ADT. It has been observed that Androgen derivation therapy combined with drugs like abiraterone acetate or docetaxel improve overall survival rate in metastatic hormone sensitive prostate cancer (mHSPC) patients. Scientific investigations have revealed that drugs inhibiting poly ADP Ribose polymerase (PARP) are showing promising results in clinical trials in the prostate cancer population with mCRPC and DNA repair abnormalities. Recently, RISUG adv (reversible inhibition of sperm under guidance) has shown significant results against prostate cancer cell lines and MTT assay has validated substantial effects of this drug against PC3 cell lines. Current review paper highlights the advancements in prostate cancer therapeutics and new drug molecules against prostate cancer. It will provide detailed insights on the signaling pathways which need to be targeted to combat metastasized prostate cancer and castration resistant prostate cancer.
Collapse
Affiliation(s)
- Neha Thakur
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand, India
| | - Sameer Quazi
- Department of Chemistry, Akshara First Grade College, Bengaluru, India
- GenLab Biosolutions Private Limited, Bangalore, Karnataka, India
- Department of Biomedical Sciences, School of Life Sciences, Anglia Ruskin University, Cambridge, United Kingdom
- School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Solution Chemistry of Advanced Materials and Technologies (SCAMT) Institute, ITMO University, St. Petersburg, Russia
| | - Bindu Naik
- Department of Food Science and Technology, Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, India
| | - Pallavi Singh
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand, India
| |
Collapse
|
21
|
Kiełb P, Kowalczyk K, Gurwin A, Nowak Ł, Krajewski W, Sosnowski R, Szydełko T, Małkiewicz B. Novel Histopathological Biomarkers in Prostate Cancer: Implications and Perspectives. Biomedicines 2023; 11:1552. [PMID: 37371647 DOI: 10.3390/biomedicines11061552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/12/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Prostate cancer (PCa) is the second most frequently diagnosed cancer in men. Despite the significant progress in cancer diagnosis and treatment over the last few years, the approach to disease detection and therapy still does not include histopathological biomarkers. The dissemination of PCa is strictly related to the creation of a premetastatic niche, which can be detected by altered levels of specific biomarkers. To date, the risk factors for biochemical recurrence include lymph node status, prostate-specific antigen (PSA), PSA density (PSAD), body mass index (BMI), pathological Gleason score, seminal vesicle invasion, extraprostatic extension, and intraductal carcinoma. In the future, biomarkers might represent another prognostic factor, as discussed in many studies. In this review, we focus on histopathological biomarkers (particularly CD169 macrophages, neuropilin-1, cofilin-1, interleukin-17, signal transducer and activator of transcription protein 3 (STAT3), LIM domain kinase 1 (LIMK1), CD15, AMACR, prostate-specific membrane antigen (PSMA), Appl1, Sortilin, Syndecan-1, and p63) and their potential application in decision making regarding the prognosis and treatment of PCa patients. We refer to studies that found a correlation between the levels of biomarkers and tumor characteristics as well as clinical outcomes. We also hypothesize about the potential use of histopathological markers as a target for novel immunotherapeutic drugs or targeted radionuclide therapy, which may be used as adjuvant therapy in the future.
Collapse
Affiliation(s)
- Paweł Kiełb
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| | - Kamil Kowalczyk
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| | - Adam Gurwin
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| | - Łukasz Nowak
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| | - Wojciech Krajewski
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| | - Roman Sosnowski
- Department of Urogenital Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Tomasz Szydełko
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| | - Bartosz Małkiewicz
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| |
Collapse
|
22
|
Lankoff A, Czerwińska M, Kruszewski M. Nanoparticle-Based Radioconjugates for Targeted Imaging and Therapy of Prostate Cancer. Molecules 2023; 28:molecules28104122. [PMID: 37241862 DOI: 10.3390/molecules28104122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/24/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Prostate cancer is the second most frequent malignancy in men worldwide and the fifth leading cause of death by cancer. Although most patients initially benefit from therapy, many of them will progress to metastatic castration-resistant prostate cancer, which still remains incurable. The significant mortality and morbidity rate associated with the progression of the disease results mainly from a lack of specific and sensitive prostate cancer screening systems, identification of the disease at mature stages, and failure of anticancer therapy. To overcome the limitations of conventional imaging and therapeutic strategies for prostate cancer, various types of nanoparticles have been designed and synthesized to selectively target prostate cancer cells without causing toxic side effects to healthy organs. The purpose of this review is to briefly discuss the selection criteria of suitable nanoparticles, ligands, radionuclides, and radiolabelling strategies for the development of nanoparticle-based radioconjugates for targeted imaging and therapy of prostate cancer and to evaluate progress in the field, focusing attention on their design, specificity, and potential for detection and/or therapy.
Collapse
Affiliation(s)
- Anna Lankoff
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland
- Department of Medical Biology, Institute of Biology, Jan Kochanowski University, Uniwersytecka 15, 25-406 Kielce, Poland
| | - Malwina Czerwińska
- Department of Dietetics, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences (WULS-SGGW), 159c Nowoursynowska, 02-776 Warsaw, Poland
| | - Marcin Kruszewski
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland
- Department of Molecular Biology and Translational Research, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland
| |
Collapse
|
23
|
Heynickx N, Segers C, Coolkens A, Baatout S, Vermeulen K. Characterization of Non-Specific Uptake and Retention Mechanisms of [ 177Lu]Lu-PSMA-617 in the Salivary Glands. Pharmaceuticals (Basel) 2023; 16:ph16050692. [PMID: 37242475 DOI: 10.3390/ph16050692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
The radionuclide therapy [177Lu]Lu-PSMA-617 was recently FDA-approved for treatment of metastatic castration-resistant prostate cancer. Salivary gland toxicity is currently considered as the main dose-limiting side effect. However, its uptake and retention mechanisms in the salivary glands remain elusive. Therefore, our aim was to elucidate the uptake patterns of [177Lu]Lu-PSMA-617 in salivary gland tissue and cells by conducting cellular binding and autoradiography experiments. Briefly, A-253 and PC3-PIP cells, and mouse kidney and pig salivary gland tissue, were incubated with 5 nM [177Lu]Lu-PSMA-617 to characterize its binding. Additionally, [177Lu]Lu-PSMA-617 was co-incubated with monosodium glutamate, ionotropic or metabotropic glutamate receptor antagonists. Low, non-specific binding was observed in salivary gland cells and tissues. Monosodium glutamate was able to decrease [177Lu]Lu-PSMA-617 in PC3-PIP cells, mouse kidney and pig salivary gland tissue. Kynurenic acid (ionotropic antagonist) decreased the binding of [177Lu]Lu-PSMA-617 to 29.2 ± 20.6% and 63.4 ± 15.4%, respectively, with similar effects observed on tissues. (RS)-MCPG (metabotropic antagonist) was able to decrease the [177Lu]Lu-PSMA-617 binding on A-253 cells to 68.2 ± 16.8% and pig salivary gland tissue to 53.1 ± 36.8%. To conclude, we showed that the non-specific binding on [177Lu]Lu-PSMA-617 could be reduced by monosodium glutamate, kynurenic acid and (RS)-MCPG.
Collapse
Affiliation(s)
- Nathalie Heynickx
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), 2400 Mol, Belgium
- Department of Molecular Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Charlotte Segers
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), 2400 Mol, Belgium
| | - Amelie Coolkens
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), 2400 Mol, Belgium
| | - Sarah Baatout
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), 2400 Mol, Belgium
- Department of Molecular Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
- Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Koen Vermeulen
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), 2400 Mol, Belgium
| |
Collapse
|
24
|
Lopes FPPL, Pedras MV, Ferreira ÁRS, Ledesma PM, Dias PRTP, Pedras FV. Behavior of total alkaline phosphatase after radium-233 therapy in metastatic castration-resistant prostate cancer: a single-center, real-world retrospective study. Radiol Bras 2023; 56:125-130. [PMID: 37564085 PMCID: PMC10411772 DOI: 10.1590/0100-3984.2022.0080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 10/27/2022] [Indexed: 08/12/2023] Open
Abstract
Objective To describe the behavior of total alkaline phosphatase (tALP) in patients with metastatic castration-resistant prostate cancer receiving radium-223 therapy, in a real-world scenario, and to describe overall survival (OS) among such patients. Materials and Methods This was a retrospective study involving 97 patients treated between February 2017 and September 2020. Patients were stratified by the baseline tALP (normal/elevated). A tALP response was defined as a ≥ 30% reduction from baseline at week 12. For patients with elevated baseline tALP, we also evaluated treatment response as a ≥ 10% reduction in tALP after the first cycle of treatment. We defined OS as the time from the first treatment cycle to the date of death. Results There was a significant reduction in the median tALP after each cycle of treatment (p < 0.05 for all). Data for tALP at week 12 were available for 71 of the 97 patients. Of those 71 patients, 26 (36.6%) responded. Elevated baseline tALP was observed in 47 patients, of whom 19 (40.4%) showed a response. Longer OS was observed in the patients with normal baseline tALP, in those with elevated baseline tALP that showed a response to treatment (≥ 10% reduction), and in those who received 5-6 cycles of therapy. Conclusion The tALP may be used to predict which patients will benefit from treatment with a greater number of cycles of radium-223 therapy and will have longer OS.
Collapse
|
25
|
Sreekumar S, Zhou D, Mpoy C, Schenk E, Scott J, Arbeit JM, Xu J, Rogers BE. Preclinical Efficacy of a PARP-1 Targeted Auger-Emitting Radionuclide in Prostate Cancer. Int J Mol Sci 2023; 24:3083. [PMID: 36834491 PMCID: PMC9967758 DOI: 10.3390/ijms24043083] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 02/09/2023] Open
Abstract
There is an unmet need for better therapeutic strategies for advanced prostate cancer. Poly (ADP-ribose) polymerase-1 (PARP-1) is a chromatin-binding DNA repair enzyme overexpressed in prostate cancer. This study evaluates whether PARP-1, on account of its proximity to the cell's DNA, would be a good target for delivering high-linear energy transfer Auger radiation to induce lethal DNA damage in prostate cancer cells. We analyzed the correlation between PARP-1 expression and Gleason score in a prostate cancer tissue microarray. A radio-brominated Auger emitting inhibitor ([77Br]Br-WC-DZ) targeting PARP-1 was synthesized. The ability of [77Br]Br-WC-DZ to induce cytotoxicity and DNA damage was assessed in vitro. The antitumor efficacy of [77Br]Br-WC-DZ was investigated in prostate cancer xenograft models. PARP-1 expression was found to be positively correlated with the Gleason score, thus making it an attractive target for Auger therapy in advanced diseases. The Auger emitter, [77Br]Br-WC-DZ, induced DNA damage, G2-M cell cycle phase arrest, and cytotoxicity in PC-3 and IGR-CaP1 prostate cancer cells. A single dose of [77Br]Br-WC-DZ inhibited the growth of prostate cancer xenografts and improved the survival of tumor-bearing mice. Our studies establish the fact that PARP-1 targeting Auger emitters could have therapeutic implications in advanced prostate cancer and provides a strong rationale for future clinical investigation.
Collapse
Affiliation(s)
- Sreeja Sreekumar
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dong Zhou
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Cedric Mpoy
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Elsa Schenk
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jalen Scott
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jeffrey M. Arbeit
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jinbin Xu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Buck E. Rogers
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
26
|
Sun JX, Xu JZ, An Y, Ma SY, Liu CQ, Zhang SH, Luan Y, Wang SG, Xia QD. Future in precise surgery: Fluorescence-guided surgery using EVs derived fluorescence contrast agent. J Control Release 2023; 353:832-841. [PMID: 36496053 DOI: 10.1016/j.jconrel.2022.12.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022]
Abstract
Surgery is the only cure for many solid tumors, but positive resection margins, damage to vital nerves, vessels and organs during surgery, and the range and extent of lymph node dissection are significant concerns which hinder the development of surgery. The emergence of fluorescence-guided surgery (FGS) means a farewell to the era when surgeons relied only on visual and tactile feedback, and it gives surgeons another eye to distinguish tumors from normal tissues for precise resection and helps to find a balance between complete tumor lesions removal and maximal organ function conservation. However, the existing synthetic fluorescence contrast agent has flaws in safety, specificity and biocompatibility to various extents. Extracellular vesicles (EVs) are a group of heterogeneous types of cell-derived membranous structures present in all biological fluids. EVs, especially engineered targeting EVs, play an increasingly important role in drug delivery because of their good biocompatibility, validated safety and targeting ability. Nevertheless, few studies have employed EVs loaded with fluorophores to construct fluorescence contrast agents and used them in FGS. Here, we systematically reviewed the current state of knowledge regarding FGS, fundamental characteristics of EVs, and the development of engineered targeting EVs, and put forward a novel strategy and procedures to produce EVs-based fluorescence contrast agent used in fluorescence-guided surgery.
Collapse
Affiliation(s)
- Jian-Xuan Sun
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, 430030 Wuhan, China
| | - Jin-Zhou Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, 430030 Wuhan, China
| | - Ye An
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, 430030 Wuhan, China
| | - Si-Yang Ma
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, 430030 Wuhan, China
| | - Chen-Qian Liu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, 430030 Wuhan, China
| | - Si-Han Zhang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, 430030 Wuhan, China
| | - Yang Luan
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, 430030 Wuhan, China.
| | - Shao-Gang Wang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, 430030 Wuhan, China.
| | - Qi-Dong Xia
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, 430030 Wuhan, China.
| |
Collapse
|
27
|
Li Q, Liu Z, Ma L, Yin W, Zhang K. CACYBP knockdown inhibits progression of prostate cancer via p53. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04497-x. [PMID: 36576589 PMCID: PMC10356884 DOI: 10.1007/s00432-022-04497-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/24/2022] [Indexed: 12/29/2022]
Abstract
PURPOSE Prostate cancer (PC) is one of the most common malignant tumors of genitourinary system in men. CACYCLIN binding protein (CACYBP) is involved in the progression of a variety of cancers. The aim of this study was to explore the expression and functional role of CACYBP in PC. METHODS The expression of CACYBP in PC was evaluated by immunohistochemical (IHC) staining and qRT-PCR. Subsequently, we established lentivirus-mediated CACYBP knockdown in PC cell lines. The biological roles of CACYBP on proliferation, apoptosis, cycle distribution, migration and tumor formation of PC were investigated by Celigo cell counting assay, flow cytometry, transwell assay, wound-healing assay and mice xenograft models, respectively. RESULTS CACYBP was highly expressed in PC and was positively correlated with the pathological grade of PC patients. Knockdown of CACYBP inhibited proliferation, enhanced apoptosis, arrested cell cycle in G2 and suppressed migration of PC cell lines in vitro. In addition, CACYBP knockdown weakened the tumor growth of PC in vivo. Moreover, addition of p53 inhibitor could effectively alleviate the inhibitory effect of CACYBP knockdown on cell activity. CONCLUSION This study revealed that knockdown of CACYBP inhibited the proliferation, migration and tumorigenicity of PC, which may serve as a potential therapeutic target for the treatment of PC.
Collapse
Affiliation(s)
- Qiang Li
- Department of Urological Surgery, First Affiliated Hospital School of Medicine, Shihezi University, Shihezi, 832008, Xinjiang, China
| | - Zhili Liu
- Department of Urological Surgery, First Affiliated Hospital School of Medicine, Shihezi University, Shihezi, 832008, Xinjiang, China
| | - Luping Ma
- Department of Urological Surgery, First Affiliated Hospital School of Medicine, Shihezi University, Shihezi, 832008, Xinjiang, China
| | - Weiqi Yin
- Department of Urological Surgery, Ningbo First Hospital, Ningbo, 315010, Zhejiang, China
| | - Kan Zhang
- Department of Urological Surgery, Ningbo First Hospital, Ningbo, 315010, Zhejiang, China.
| |
Collapse
|
28
|
Intercomparison of S-Factor values calculated in Zubal voxelized phantom for eleven radionuclides commonly used in targeted prostate cancer therapy. Phys Eng Sci Med 2022; 45:1251-1256. [PMID: 36315382 DOI: 10.1007/s13246-022-01191-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/20/2022] [Indexed: 12/14/2022]
Abstract
In this study we aimed at comparing various radionuclides ordinarily used in targeted prostate cancer therapy, thereby evaluating S-Factor parameter in the prostate organs as well as in its surrounding healthy tissues, namely the urinary bladder and rectum. InterDosi code version 1.1 was used to estimate S-Factor values in Zubal voxelized phantom for 11 radionuclides, namely 225Ac, 21At, 67Cu, 125I, 131I, 212Pb, 177Lu, 223Ra, 161 Tb, 227Th and 90Y. The prostate organ was considered the source of different ionizing radiation emitted by the radionuclides cited above. The results showed that among all studied alpha-emitting radionuclides, 225 Ac, 223 Ra and 227 Th provide equidistantly the highest self-irradiation S-Factors whereas, 211At provides the lowest cross-irradiation S-Factors. On the other hand, considering only beta-emitting radionuclides, it is shown that 177Lu and 90Y induce respectively lowest and highest cross-absorption S-Factors on the surrounding healthy organs. We conclude that 177Lu and 211At are more adequate for prostate radionuclide therapy because they can relatively prevent surrounding organs from radiation toxicity and at the same time provide sufficient dose to treat the prostate tumor.
Collapse
|
29
|
Poulie CBM, Sporer E, Hvass L, Jørgensen JT, Kempen PJ, Lopes van den Broek SI, Shalgunov V, Kjaer A, Jensen AI, Herth MM. Bioorthogonal Click of Colloidal Gold Nanoparticles to Antibodies In vivo. Chemistry 2022; 28:e202201847. [PMID: 35851967 DOI: 10.1002/chem.202201847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Indexed: 01/07/2023]
Abstract
Combining nanotechnology and bioorthogonal chemistry for theranostic strategies offers the possibility to develop next generation nanomedicines. These materials are thought to increase therapeutic outcome and improve current cancer management. Due to their size, nanomedicines target tumors passively. Thus, they can be used for drug delivery purposes. Bioorthogonal chemistry allows for a pretargeting approach. Higher target-to-background drug accumulation ratios can be achieved. Pretargeting can also be used to induce internalization processes or trigger controlled drug release. Colloidal gold nanoparticles (AuNPs) have attracted widespread interest as drug delivery vectors within the last decades. Here, we demonstrate for the first time the possibility to successfully ligate AuNPs in vivo to pretargeted monoclonal antibodies. We believe that this possibility will facilitate the development of AuNPs for clinical use and ultimately, improve state-of-the-art patient care.
Collapse
Affiliation(s)
- Christian B M Poulie
- Department of Drug Design and Pharmacology Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100, Copenhagen, Denmark
| | - Emanuel Sporer
- Center for Nanomedicine and Theranostics, DTU Health Technology, Technical University of Denmark (DTU), Ørsteds Plads 345C, 2800, Lyngby, Denmark
| | - Lars Hvass
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark.,Cluster for Molecular Imaging Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Jesper T Jørgensen
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark.,Cluster for Molecular Imaging Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Paul J Kempen
- National Centre for Nano Fabrication and Characterization, Technical University of Denmark (DTU), Ørsteds Plads 347, 2800, Lyngby, Denmark
| | - Sara I Lopes van den Broek
- Department of Drug Design and Pharmacology Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100, Copenhagen, Denmark
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100, Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark.,Cluster for Molecular Imaging Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Andreas I Jensen
- Center for Nanomedicine and Theranostics, DTU Health Technology, Technical University of Denmark (DTU), Ørsteds Plads 345C, 2800, Lyngby, Denmark
| | - Matthias M Herth
- Department of Drug Design and Pharmacology Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100, Copenhagen, Denmark.,Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| |
Collapse
|
30
|
Kara A. An optimization study for targeted alpha therapy: Ion behaviours and dose calculations within ICRU-compact bone tissue. Appl Radiat Isot 2022; 191:110552. [DOI: 10.1016/j.apradiso.2022.110552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/27/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
|
31
|
Sun J, Huangfu Z, Yang J, Wang G, Hu K, Gao M, Zhong Z. Imaging-guided targeted radionuclide tumor therapy: From concept to clinical translation. Adv Drug Deliv Rev 2022; 190:114538. [PMID: 36162696 DOI: 10.1016/j.addr.2022.114538] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 09/03/2022] [Accepted: 09/11/2022] [Indexed: 01/24/2023]
Abstract
Since the first introduction of sodium iodide I-131 for use with thyroid patients almost 80 years ago, more than 50 radiopharmaceuticals have reached the markets for a wide range of diseases, especially cancers. The nuclear medicine paradigm also shifts from solely molecular imaging or radionuclide therapy to imaging-guided radionuclide therapy, which is deemed a vital component of precision cancer therapy and an emerging medical modality for personalized medicine. The imaging-guided radionuclide therapy highlights the systematic integration of targeted nuclear diagnostics and radionuclide therapeutics. Regarding this, nuclear imaging serves to "visualize" the lesions and guide the therapeutic strategy, followed by administration of a precise patient specific dose of radiotherapeutics for treatment according to the absorbed dose to different organs and tumors calculated by dosimetry tools, and finally repeated imaging to predict the prognosis. This strategy leads to significantly enhanced therapeutic efficacy, improved patient outcomes, and manageable adverse events. In this review, we provide an overview of imaging-guided targeted radionuclide therapy for different tumors such as advanced prostate cancer and neuroendocrine tumors, with a focus on development of new radioligands and their preclinical and clinical results, and further discuss about challenges and future perspectives.
Collapse
Affiliation(s)
- Juan Sun
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhenyuan Huangfu
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Jiangtao Yang
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Guanglin Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China.
| | - Kuan Hu
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan.
| | - Mingyuan Gao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhiyuan Zhong
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China.
| |
Collapse
|
32
|
Chen J, Qi L, Tang Y, Tang G, Gan Y, Cai Y. Current role of prostate-specific membrane antigen-based imaging and radioligand therapy in castration-resistant prostate cancer. Front Cell Dev Biol 2022; 10:958180. [PMID: 36036001 PMCID: PMC9411749 DOI: 10.3389/fcell.2022.958180] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/11/2022] [Indexed: 11/29/2022] Open
Abstract
Castration-resistant prostate cancer (CRPC) is a therapy-resistant and lethal form of prostate cancer as well as a therapeutic challenge. Prostate-specific membrane antigen (PSMA) has been proved as a promising molecular target for optimizing the theranostics for CRPC patients. When combined with PSMA radiotracers, novel molecular imaging techniques such as positron emission tomography (PET) can provide more accurate and expedient identification of metastases when compared with conventional imaging techniques. Based on the PSMA-based PET scans, the accurate visualization of local and disseminative lesions may help in metastasis-directed therapy. Moreover, the combination of 68Ga-labeled PSMA-based PET imaging and radiotherapy using PSMA radioligand therapy (RLT) becomes a novel treatment option for CRPC patients. The existing studies have demonstrated this therapeutic strategy as an effective and well-tolerated therapy among CRPC patients. PSMA-based PET imaging can accurately detect CRPC lesions and describe their molecular features with quantitative parameters, which can be used to select the best choice of treatments, monitor the response, and predict the outcome of RLT. This review discussed the current and potential role of PSMA‐based imaging and RLT in the diagnosis, treatment, and prediction of prognosis of CRPC.
Collapse
Affiliation(s)
- Jiaxian Chen
- Department of Urology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Lin Qi
- Department of Urology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yongxiang Tang
- Department of PET Center, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Guyu Tang
- Department of Urology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yu Gan
- Department of Urology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- *Correspondence: Yu Gan, ; Yi Cai,
| | - Yi Cai
- Department of Urology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- *Correspondence: Yu Gan, ; Yi Cai,
| |
Collapse
|
33
|
Ruigrok EAM, Verkaik NS, de Blois E, de Ridder C, Stuurman D, Roobol SJ, Van Gent DC, de Jong M, Van Weerden WM, Nonnekens J. Preclinical Assessment of the Combination of PSMA-Targeting Radionuclide Therapy with PARP Inhibitors for Prostate Cancer Treatment. Int J Mol Sci 2022; 23:ijms23148037. [PMID: 35887398 PMCID: PMC9316488 DOI: 10.3390/ijms23148037] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/12/2022] [Accepted: 07/18/2022] [Indexed: 01/22/2023] Open
Abstract
Prostate specific membrane antigen targeted radionuclide therapy (PSMA-TRT) is a promising novel treatment for prostate cancer (PCa) patients. However, PSMA-TRT cannot be used for curative intent yet, thus additional research on how to improve the therapeutic efficacy is warranted. A potential way of achieving this, is combining TRT with poly ADP-ribosylation inhibitors (PARPi), which has shown promising results for TRT of neuroendocrine tumor cells. Currently, several clinical trials have been initiated for this combination for PCa, however so far, no evidence of synergism is available for PCa. Therefore, we evaluated the combination of PSMA-TRT with three classes of PARPi in preclinical PCa models. In vitro viability and survival assays were performed using PSMA-expressing PCa cell lines PC3-PIP and LNCaP to assess the effect of increasing concentrations of PARPi veliparib, olaparib or talazoparib in combination with PSMA-TRT compared to single PARPi treatment. Next, DNA damage analyses were performed by quantifying the number of DNA breaks by immunofluorescent stainings. Lastly, the potential of the combination treatments was studied in vivo in mice bearing PC3-PIP xenografts. Our results show that combining PSMA-TRT with PARPi did not synergistically affect the in vitro clonogenic survival or cell viability. DNA-damage analysis revealed only a significant increase in DNA breaks when combining PSMA-TRT with veliparib and not in the other combination treatments. Moreover, PSMA-TRT with PARPi treatment did not improve tumor control compared to PSMA-TRT monotherapy. Overall, the data presented do not support the assumption that combining PSMA-TRT with PARPi leads to a synergistic antitumor effect in PCa. These results underline that extensive preclinical research using various PCa models is imperative to validate the applicability of the combination strategy for PCa, as it is for other cancer types.
Collapse
Affiliation(s)
- Eline A. M. Ruigrok
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (E.A.M.R.); (E.d.B.); (C.d.R.); (D.S.); (S.J.R.); (M.d.J.)
- Department of Experimental Urology, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands;
| | - Nicole S. Verkaik
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (N.S.V.); (D.C.V.G.)
| | - Erik de Blois
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (E.A.M.R.); (E.d.B.); (C.d.R.); (D.S.); (S.J.R.); (M.d.J.)
| | - Corrina de Ridder
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (E.A.M.R.); (E.d.B.); (C.d.R.); (D.S.); (S.J.R.); (M.d.J.)
- Department of Experimental Urology, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands;
| | - Debra Stuurman
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (E.A.M.R.); (E.d.B.); (C.d.R.); (D.S.); (S.J.R.); (M.d.J.)
- Department of Experimental Urology, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands;
| | - Stefan J. Roobol
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (E.A.M.R.); (E.d.B.); (C.d.R.); (D.S.); (S.J.R.); (M.d.J.)
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (N.S.V.); (D.C.V.G.)
| | - Dik C. Van Gent
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (N.S.V.); (D.C.V.G.)
| | - Marion de Jong
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (E.A.M.R.); (E.d.B.); (C.d.R.); (D.S.); (S.J.R.); (M.d.J.)
| | - Wytske M. Van Weerden
- Department of Experimental Urology, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands;
| | - Julie Nonnekens
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (E.A.M.R.); (E.d.B.); (C.d.R.); (D.S.); (S.J.R.); (M.d.J.)
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (N.S.V.); (D.C.V.G.)
- Correspondence:
| |
Collapse
|
34
|
In vitro dose effect relationships of actinium-225- and lutetium-177-labeled PSMA-I&T. Eur J Nucl Med Mol Imaging 2022; 49:3627-3638. [PMID: 35556158 PMCID: PMC9399067 DOI: 10.1007/s00259-022-05821-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 04/25/2022] [Indexed: 12/18/2022]
Abstract
PURPOSE Targeting the prostate-specific membrane antigen (PSMA) using lutetium-177-labeled PSMA-specific tracers has become a very promising novel therapy option for prostate cancer (PCa). The efficacy of this therapy might be further improved by replacing the β-emitting lutetium-177 with the α-emitting actinium-225. Actinium-225 is thought to have a higher therapeutic efficacy due to the high linear energy transfer (LET) of the emitted α-particles, which can increase the amount and complexity of the therapy induced DNA double strand breaks (DSBs). Here we evaluated the relative biological effectiveness of [225Ac]Ac-PSMA-I&T and [177Lu]Lu-PSMA-I&T by assessing in vitro binding characteristics, dosimetry, and therapeutic efficacy. METHODS AND RESULTS The PSMA-expressing PCa cell line PC3-PIP was used for all in vitro assays. First, binding and displacement assays were performed, which revealed similar binding characteristics between [225Ac]Ac-PSMA-I&T and [177Lu]Lu-PSMA-I&T. Next, the assessment of the number of 53BP1 foci, a marker for the number of DNA double strand breaks (DSBs), showed that cells treated with [225Ac]Ac-PSMA-I&T had slower DSB repair kinetics compared to cells treated with [177Lu]Lu-PSMA-I&T. Additionally, clonogenic survival assays showed that specific targeting with [225Ac]Ac-PSMA-I&T and [177Lu]Lu-PSMA-I&T caused a dose-dependent decrease in survival. Lastly, after dosimetric assessment, the relative biological effectiveness (RBE) of [225Ac]Ac-PSMA-I&T was found to be 4.2 times higher compared to [177Lu]Lu-PSMA-I&T. CONCLUSION We found that labeling of PSMA-I&T with lutetium-177 or actinium-225 resulted in similar in vitro binding characteristics, indicating that the distinct biological effects observed in this study are not caused by a difference in uptake of the two tracers. The slower repair kinetics of [225Ac]Ac-PSMA-I&T compared to [177Lu]Lu-PSMA-I&T correlates to the assumption that irradiation with actinium-225 causes more complex, more difficult to repair DSBs compared to lutetium-177 irradiation. Furthermore, the higher RBE of [225Ac]Ac-PSMA-I&T compared to [177Lu]Lu-PSMA-I&T underlines the therapeutic potential for the treatment of PCa.
Collapse
|
35
|
Halib N, Pavan N, Trombetta C, Dapas B, Farra R, Scaggiante B, Grassi M, Grassi G. An Overview of siRNA Delivery Strategies for Urological Cancers. Pharmaceutics 2022; 14:pharmaceutics14040718. [PMID: 35456552 PMCID: PMC9030829 DOI: 10.3390/pharmaceutics14040718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/18/2022] [Accepted: 03/24/2022] [Indexed: 02/05/2023] Open
Abstract
The treatment of urological cancers has been significantly improved in recent years. However, for the advanced stages of these cancers and/or for those developing resistance, novel therapeutic options need to be developed. Among the innovative strategies, the use of small interfering RNA (siRNA) seems to be of great therapeutic interest. siRNAs are double-stranded RNA molecules which can specifically target virtually any mRNA of pathological genes. For this reason, siRNAs have a great therapeutic potential for human diseases including urological cancers. However, the fragile nature of siRNAs in the biological environment imposes the development of appropriate delivery systems to protect them. Thus, ensuring siRNA reaches its deep tissue target while maintaining structural and functional integrity represents one of the major challenges. To reach this goal, siRNA-based therapies require the development of fine, tailor-made delivery systems. Polymeric nanoparticles, lipid nanoparticles, nanobubbles and magnetic nanoparticles are among nano-delivery systems studied recently to meet this demand. In this review, after an introduction about the main features of urological tumors, we describe siRNA characteristics together with representative delivery systems developed for urology applications; the examples reported are subdivided on the basis of the different delivery materials and on the different urological cancers.
Collapse
Affiliation(s)
- Nadia Halib
- Department of Basic Sciences & Oral Biology, Faculty of Dentistry, Universiti Sains Islam Malaysia, Kuala Lumpur 55100, Malaysia;
| | - Nicola Pavan
- Urology Clinic, Department of Medical, Surgical and Health Science, University of Trieste, I-34149 Trieste, Italy; (N.P.); (C.T.)
| | - Carlo Trombetta
- Urology Clinic, Department of Medical, Surgical and Health Science, University of Trieste, I-34149 Trieste, Italy; (N.P.); (C.T.)
| | - Barbara Dapas
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I-34149 Trieste, Italy; (B.D.); (R.F.); (B.S.)
| | - Rossella Farra
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I-34149 Trieste, Italy; (B.D.); (R.F.); (B.S.)
| | - Bruna Scaggiante
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I-34149 Trieste, Italy; (B.D.); (R.F.); (B.S.)
| | - Mario Grassi
- Department of Engineering and Architecture, Trieste University, Via Valerio 6, I-34127 Trieste, Italy;
| | - Gabriele Grassi
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I-34149 Trieste, Italy; (B.D.); (R.F.); (B.S.)
- Correspondence: ; Tel.: +39-040-399-3227
| |
Collapse
|
36
|
Wang F, Li Z, Feng X, Yang D, Lin M. Advances in PSMA-targeted therapy for prostate cancer. Prostate Cancer Prostatic Dis 2022; 25:11-26. [PMID: 34050265 DOI: 10.1038/s41391-021-00394-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/27/2021] [Accepted: 05/12/2021] [Indexed: 02/04/2023]
Abstract
Prostate-specific membrane antigen (PSMA), a transmembrane glycoprotein located on the cell membrane, is specifically and highly expressed in prostate cancer (PCa). Besides, its expression level is related to tumor invasiveness. As a molecular target of PCa, PSMA has been extensively studied in the past two decades. Currently, a great deal of evidence suggests that significant progresses have been made in the PSMA-targeted therapy of PCa. Herein, different PSMA-targeted therapies for PCa are reviewed, including radioligand therapy (177Lu-PSMA-RLT, 225Ac-PSMA-RLT), antibody-drug conjugates (MLN2704, PSMA-MMAE, MEDI3726), cellular immunotherapy (CAR-T, CAR/NK-92, PSMA-targeted BiTE), photodynamic therapy, imaging-guided surgery (radionuclide-guided surgery, fluorescence-guided surgery, multimodal imaging-guided surgery), and ultrasound-mediated nanobubble destruction.
Collapse
Affiliation(s)
- Fujin Wang
- Nantong University, Nantong, Jiangsu, China.,Department of Radiology, the First People's Hospital of Yancheng, Yancheng, Jiangsu, China
| | - Zhifeng Li
- Nantong University, Nantong, Jiangsu, China
| | - Xiaoqian Feng
- Nantong University, Nantong, Jiangsu, China.,Department of Radiology, the First People's Hospital of Yancheng, Yancheng, Jiangsu, China
| | | | - Mei Lin
- Taizhou People's Hospital Affiliated to Nantong University, Taizhou, Jiangsu, China.
| |
Collapse
|
37
|
Seniwal B, Thipe VC, Singh S, Fonseca TCF, Freitas de Freitas L. Recent Advances in Brachytherapy Using Radioactive Nanoparticles: An Alternative to Seed-Based Brachytherapy. Front Oncol 2021; 11:766407. [PMID: 34900715 PMCID: PMC8651618 DOI: 10.3389/fonc.2021.766407] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 10/29/2021] [Indexed: 12/24/2022] Open
Abstract
Interstitial brachytherapy (BT) is generally used for the treatment of well-confined solid tumors. One example of this is in the treatment of prostate tumors by permanent placement of radioactive seeds within the prostate gland, where low doses of radiation are delivered for several months. However, successful implementation of this technique is hampered due to several posttreatment adverse effects or symptoms and operational and logistical complications associated with it. Recently, with the advancements in nanotechnology, radioactive nanoparticles (radio-NPs) functionalized with tumor-specific biomolecules, injected intratumorally, have been reported as an alternative to seed-based BT. Successful treatment of solid tumors using radio-NPs has been reported in several preclinical studies, on both mice and canine models. In this article, we review the recent advancements in the synthesis and use of radio-NPs as a substitute to seed-based BT. Here, we discuss the limitations of current seed-based BT and advantages of radio-NPs for BT applications. Recent progress on the types of radio-NPs, their features, synthesis methods, and delivery techniques are discussed. The last part of the review focuses on the currently used dosimetry protocols and studies on the dosimetry of nanobrachytherapy applications using radio-NPs. The current challenges and future research directions on the role of radio-NPs in BT treatments are also discussed.
Collapse
Affiliation(s)
- Baljeet Seniwal
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval (CR-CHU de Québec), Axe Médecine Régénératrice, Québec, QC, Canada
| | - Velaphi C Thipe
- Instituto de Pesquisas Energéticas e Nucleares, Comissão Nacional de Energia Nuclear (IPEN-CNEN), Cidade Universitária, São Paulo, Brazil.,Department of Radiology, Institute of Green Nanotechnology, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Sukhvir Singh
- Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organisation, Delhi, India
| | - Telma C F Fonseca
- Departamento de Engenharia Nuclear-Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lucas Freitas de Freitas
- Instituto de Pesquisas Energéticas e Nucleares, Comissão Nacional de Energia Nuclear (IPEN-CNEN), Cidade Universitária, São Paulo, Brazil
| |
Collapse
|
38
|
Freitas LF, Ferreira AH, Thipe VC, Varca GHC, Lima CSA, Batista JGS, Riello FN, Nogueira K, Cruz CPC, Mendes GOA, Rodrigues AS, Sousa TS, Alves VM, Lugão AB. The State of the Art of Theranostic Nanomaterials for Lung, Breast, and Prostate Cancers. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:2579. [PMID: 34685018 PMCID: PMC8539690 DOI: 10.3390/nano11102579] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/14/2021] [Accepted: 09/24/2021] [Indexed: 02/06/2023]
Abstract
The synthesis and engineering of nanomaterials offer more robust systems for the treatment of cancer, with technologies that combine therapy with imaging diagnostic tools in the so-called nanotheranostics. Among the most studied systems, there are quantum dots, liposomes, polymeric nanoparticles, inorganic nanoparticles, magnetic nanoparticles, dendrimers, and gold nanoparticles. Most of the advantages of nanomaterials over the classic anticancer therapies come from their optimal size, which prevents the elimination by the kidneys and enhances their permeation in the tumor due to the abnormal blood vessels present in cancer tissues. Furthermore, the drug delivery and the contrast efficiency for imaging are enhanced, especially due to the increased surface area and the selective accumulation in the desired tissues. This property leads to the reduced drug dose necessary to exert the desired effect and for a longer action within the tumor. Finally, they are made so that there is no degradation into toxic byproducts and have a lower immune response triggering. In this article, we intend to review and discuss the state-of-the-art regarding the use of nanomaterials as therapeutic and diagnostic tools for lung, breast, and prostate cancer, as they are among the most prevalent worldwide.
Collapse
Affiliation(s)
- Lucas F. Freitas
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Sao Paulo 05508-000, Brazil; (A.H.F.); (V.C.T.); (C.S.A.L.); (J.G.S.B.); (F.N.R.); (K.N.); (C.P.C.C.); (G.O.A.M.); (A.S.R.); (T.S.S.); (V.M.A.); (A.B.L.)
| | - Aryel H. Ferreira
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Sao Paulo 05508-000, Brazil; (A.H.F.); (V.C.T.); (C.S.A.L.); (J.G.S.B.); (F.N.R.); (K.N.); (C.P.C.C.); (G.O.A.M.); (A.S.R.); (T.S.S.); (V.M.A.); (A.B.L.)
- MackGraphe-Graphene and Nanomaterial Research Center, Mackenzie Presbyterian University, Sao Paulo 01302-907, Brazil
| | - Velaphi C. Thipe
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Sao Paulo 05508-000, Brazil; (A.H.F.); (V.C.T.); (C.S.A.L.); (J.G.S.B.); (F.N.R.); (K.N.); (C.P.C.C.); (G.O.A.M.); (A.S.R.); (T.S.S.); (V.M.A.); (A.B.L.)
| | - Gustavo H. C. Varca
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Sao Paulo 05508-000, Brazil; (A.H.F.); (V.C.T.); (C.S.A.L.); (J.G.S.B.); (F.N.R.); (K.N.); (C.P.C.C.); (G.O.A.M.); (A.S.R.); (T.S.S.); (V.M.A.); (A.B.L.)
| | - Caroline S. A. Lima
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Sao Paulo 05508-000, Brazil; (A.H.F.); (V.C.T.); (C.S.A.L.); (J.G.S.B.); (F.N.R.); (K.N.); (C.P.C.C.); (G.O.A.M.); (A.S.R.); (T.S.S.); (V.M.A.); (A.B.L.)
| | - Jorge G. S. Batista
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Sao Paulo 05508-000, Brazil; (A.H.F.); (V.C.T.); (C.S.A.L.); (J.G.S.B.); (F.N.R.); (K.N.); (C.P.C.C.); (G.O.A.M.); (A.S.R.); (T.S.S.); (V.M.A.); (A.B.L.)
| | - Fabiane N. Riello
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Sao Paulo 05508-000, Brazil; (A.H.F.); (V.C.T.); (C.S.A.L.); (J.G.S.B.); (F.N.R.); (K.N.); (C.P.C.C.); (G.O.A.M.); (A.S.R.); (T.S.S.); (V.M.A.); (A.B.L.)
| | - Kamila Nogueira
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Sao Paulo 05508-000, Brazil; (A.H.F.); (V.C.T.); (C.S.A.L.); (J.G.S.B.); (F.N.R.); (K.N.); (C.P.C.C.); (G.O.A.M.); (A.S.R.); (T.S.S.); (V.M.A.); (A.B.L.)
| | - Cassia P. C. Cruz
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Sao Paulo 05508-000, Brazil; (A.H.F.); (V.C.T.); (C.S.A.L.); (J.G.S.B.); (F.N.R.); (K.N.); (C.P.C.C.); (G.O.A.M.); (A.S.R.); (T.S.S.); (V.M.A.); (A.B.L.)
| | - Giovanna O. A. Mendes
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Sao Paulo 05508-000, Brazil; (A.H.F.); (V.C.T.); (C.S.A.L.); (J.G.S.B.); (F.N.R.); (K.N.); (C.P.C.C.); (G.O.A.M.); (A.S.R.); (T.S.S.); (V.M.A.); (A.B.L.)
| | - Adriana S. Rodrigues
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Sao Paulo 05508-000, Brazil; (A.H.F.); (V.C.T.); (C.S.A.L.); (J.G.S.B.); (F.N.R.); (K.N.); (C.P.C.C.); (G.O.A.M.); (A.S.R.); (T.S.S.); (V.M.A.); (A.B.L.)
| | - Thayna S. Sousa
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Sao Paulo 05508-000, Brazil; (A.H.F.); (V.C.T.); (C.S.A.L.); (J.G.S.B.); (F.N.R.); (K.N.); (C.P.C.C.); (G.O.A.M.); (A.S.R.); (T.S.S.); (V.M.A.); (A.B.L.)
| | - Victoria M. Alves
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Sao Paulo 05508-000, Brazil; (A.H.F.); (V.C.T.); (C.S.A.L.); (J.G.S.B.); (F.N.R.); (K.N.); (C.P.C.C.); (G.O.A.M.); (A.S.R.); (T.S.S.); (V.M.A.); (A.B.L.)
| | - Ademar B. Lugão
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Sao Paulo 05508-000, Brazil; (A.H.F.); (V.C.T.); (C.S.A.L.); (J.G.S.B.); (F.N.R.); (K.N.); (C.P.C.C.); (G.O.A.M.); (A.S.R.); (T.S.S.); (V.M.A.); (A.B.L.)
| |
Collapse
|
39
|
Immunohistochemical Reactivity of Prostate-Specific Membrane Antigen in Salivary Gland Tumors. Head Neck Pathol 2021; 16:427-433. [PMID: 34420181 PMCID: PMC9187811 DOI: 10.1007/s12105-021-01376-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/16/2021] [Indexed: 10/20/2022]
Abstract
Prostate-specific membrane antigen (PSMA) is a transmembrane glycoprotein that is overexpressed in the prostate gland and prostate cancer. PSMA has been recently used in positron emission tomography/computed tomography (PET/CT) imaging and targeted alpha-radiation therapy (TAT) for prostate cancer. Recently, the tubarial gland, a type of minor salivary gland that is described as a new organ situated in the pharynx, is reported to express PMSA. Here, we studied the expression of PSMA in common benign and malignant salivary gland tumors. We performed immunohistochemistry for PSMA in 55 salivary gland tumors comprising 10 pleomorphic adenomas, 10 Warthin tumors, 9 basal cell adenomas, 9 adenoid cystic carcinomas, 9 mucoepidermoid carcinomas, and 8 salivary duct carcinomas. PSMA was expressed in 97% of benign tumors and 77% of malignant tumors. Moreover, PSMA was expressed in 59% of normal salivary glands adjacent to the tumor. PSMA is relatively expressed in salivary gland tumors and salivary glands. Therefore, salivary gland neoplasm, and normal salivary gland, possibly demonstrate the accumulation of PSMA in PET/CT. Thus, we need to monitor the side effects in the salivary glands during TAT for prostate cancer.
Collapse
|
40
|
El Fakiri M, Geis NM, Ayada N, Eder M, Eder AC. PSMA-Targeting Radiopharmaceuticals for Prostate Cancer Therapy: Recent Developments and Future Perspectives. Cancers (Basel) 2021; 13:cancers13163967. [PMID: 34439121 PMCID: PMC8393521 DOI: 10.3390/cancers13163967] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary One of the most frequently diagnosed cancer in men is adenocarcinoma of the prostate. Once the disease is metastatic, only very limited treatment options are available, resulting in a very short median survival time of 13 months; however, this reality is gradually changing due to the discovery of prostate-specific membrane antigen (PSMA), a protein that is present in cancerous prostate tissue. Researchers have developed pharmaceuticals specific for PSMA, ranging from antibodies (mAb) to low-molecular weight molecules coupled to beta minus and alpha-emitting radionuclides for their use in targeted radionuclide therapy (TRT). TRT offers the possibility of selectively removing cancer tissue via the emission of radiation or radioactive particles within the tumour. In this article, the major milestones in PSMA ligand research and the therapeutic developments are summarised, together with a future perspective on the enhancement of current therapeutic approaches. Abstract Prostate cancer (PC) is the second most common cancer among men, with 1.3 million yearly cases worldwide. Among those cancer-afflicted men, 30% will develop metastases and some will progress into metastatic castration-resistant prostate cancer (mCRPC), which is associated with a poor prognosis and median survival time that ranges from nine to 13 months. Nevertheless, the discovery of prostate specific membrane antigen (PSMA), a marker overexpressed in the majority of prostatic cancerous tissue, revolutionised PC care. Ever since, PSMA-targeted radionuclide therapy has gained remarkable international visibility in translational oncology. Furthermore, on first clinical application, it has shown significant influence on therapeutic management and patient care in metastatic and hormone-refractory prostate cancer, a disease that previously had remained immedicable. In this article, we provide a general overview of the main milestones in the development of ligands for PSMA-targeted radionuclide therapy, ranging from the firstly developed monoclonal antibodies to the current state-of-the-art low molecular weight entities conjugated with various radionuclides, as well as potential future efforts related to PSMA-targeted radionuclide therapy.
Collapse
Affiliation(s)
- Mohamed El Fakiri
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany; (M.E.F.); (N.M.G.); (N.A.); (A.-C.E.)
- Division of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Nicolas M. Geis
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany; (M.E.F.); (N.M.G.); (N.A.); (A.-C.E.)
- Division of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Nawal Ayada
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany; (M.E.F.); (N.M.G.); (N.A.); (A.-C.E.)
- Division of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Matthias Eder
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany; (M.E.F.); (N.M.G.); (N.A.); (A.-C.E.)
- Division of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-761-270-74220
| | - Ann-Christin Eder
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany; (M.E.F.); (N.M.G.); (N.A.); (A.-C.E.)
- Division of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
41
|
Hammer S, Schlicker A, Zitzmann-Kolbe S, Baumgart S, Hagemann UB, Scholz A, Haendler B, Lejeune P, Karlsson J, Ellingsen C, Hennekes H, Nielsen CH, Juul MU, Mumberg D, Schatz CA. Darolutamide Potentiates the Antitumor Efficacy of a PSMA-targeted Thorium-227 Conjugate by a Dual Mode of Action in Prostate Cancer Models. Clin Cancer Res 2021; 27:4367-4378. [PMID: 34035067 PMCID: PMC9401501 DOI: 10.1158/1078-0432.ccr-21-0342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/23/2021] [Accepted: 05/21/2021] [Indexed: 01/07/2023]
Abstract
PURPOSE Androgen receptor (AR) inhibitors are well established in the treatment of castration-resistant prostate cancer and have recently shown efficacy also in castration-sensitive prostate cancer. Although most patients respond well to initial therapy, resistance eventually develops, and thus, more effective therapeutic approaches are needed. Prostate-specific membrane antigen (PSMA) is highly expressed in prostate cancer and presents an attractive target for radionuclide therapy. Here, we evaluated the efficacy and explored the mode of action of the PSMA-targeted thorium-227 conjugate (PSMA-TTC) BAY 2315497, an antibody-based targeted alpha-therapy, in combination with the AR inhibitor darolutamide. EXPERIMENTAL DESIGN The in vitro and in vivo antitumor efficacy and mode of action of the combination treatment were investigated in preclinical cell line-derived and patient-derived prostate cancer xenograft models with different levels of PSMA expression. RESULTS Darolutamide induced the expression of PSMA in androgen-sensitive VCaP and LNCaP cells in vitro, and the efficacy of darolutamide in combination with PSMA-TTC was synergistic in these cells. In vivo, the combination treatment showed synergistic antitumor efficacy in the low PSMA-expressing VCaP and in the high PSMA-expressing ST1273 prostate cancer models, and enhanced efficacy in the enzalutamide-resistant KUCaP-1 model. The treatments were well tolerated. Mode-of-action studies revealed that darolutamide induced PSMA expression, resulting in higher tumor uptake of PSMA-TTC, and consequently, higher antitumor efficacy, and impaired PSMA-TTC-mediated induction of DNA damage repair genes, potentially contributing to increased DNA damage. CONCLUSIONS These results provide a strong rationale to investigate PSMA-TTC in combination with AR inhibitors in patients with prostate cancer.
Collapse
Affiliation(s)
| | | | | | - Simon Baumgart
- Bayer U.S. LLC, Pharmaceuticals, Cambridge, Massachusetts
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Ng TSC, Gao X, Salari K, Zlatev DV, Heidari P, Kamran SC. Incorporating PSMA-Targeting Theranostics Into Personalized Prostate Cancer Treatment: a Multidisciplinary Perspective. Front Oncol 2021; 11:722277. [PMID: 34395293 PMCID: PMC8355555 DOI: 10.3389/fonc.2021.722277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/12/2021] [Indexed: 01/12/2023] Open
Abstract
Recent developments in prostate-specific membrane antigen (PSMA) targeted diagnostic imaging and therapeutics (theranostics) promise to advance the management of primary, biochemically recurrent, and metastatic prostate cancer. In order to maximize the clinical impact of PSMA-targeted theranostics, a coordinated approach between the clinical stakeholders involved in prostate cancer management is required. Here, we present a vision for multidisciplinary use of PSMA theranostics from the viewpoints of nuclear radiology, medical oncology, urology, and radiation oncology. We review the currently available and forthcoming PSMA-based imaging and therapeutics and examine current and potential impacts on prostate cancer management from early localized disease to advanced treatment-refractory disease. Finally, we highlight the clinical and research opportunities related to PSMA-targeted theranostics and describe the importance of multidisciplinary collaboration in this space.
Collapse
Affiliation(s)
- Thomas S C Ng
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Xin Gao
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Keyan Salari
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Dimitar V Zlatev
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Pedram Heidari
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Sophia C Kamran
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
43
|
Heynickx N, Herrmann K, Vermeulen K, Baatout S, Aerts A. The salivary glands as a dose limiting organ of PSMA- targeted radionuclide therapy: A review of the lessons learnt so far. Nucl Med Biol 2021; 98-99:30-39. [PMID: 34020337 DOI: 10.1016/j.nucmedbio.2021.04.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/25/2021] [Accepted: 04/28/2021] [Indexed: 12/24/2022]
Abstract
At present, prostate cancer remains the second most occurring cancer in men, in Europe. Treatment efficacy for therapy of advanced metastatic disease, and metastatic castration-resistant prostate cancer in particular is limited. Prostate-specific membrane antigen (PSMA) is a promising therapeutic target in prostate cancer, seeing the high amount of overexpression on prostate cancer cells. Clinical investigation of PSMA-targeted radionuclide therapy has shown good clinical efficacy. However, adverse effects are observed of which salivary gland hypofunction and xerostomia are among the most prominent. Salivary gland toxicity is currently the dose-limiting side effect for PSMA-targeted radionuclide therapy, and more specifically for PSMA-targeted alpha therapy. To date, mechanisms underlying the salivary gland uptake of PSMA-targeting compounds and the subsequent damage to the salivary glands remain largely unknown. Furthermore, preventive strategies for salivary gland uptake or strategies for treatment of salivary gland toxicity are needed. This review focuses on the current knowledge on uptake mechanisms of PSMA-targeting compounds in the salivary glands and the research performed to investigate different strategies to prevent or treat salivary gland toxicity.
Collapse
Affiliation(s)
- Nathalie Heynickx
- Institute for Environment, Health and Safety, Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium; Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| | - Ken Herrmann
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America; Department of Nuclear Medicine, Medical Faculty, University Hospital Essen, Essen, Germany
| | - Koen Vermeulen
- Institute for Environment, Health and Safety, Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Sarah Baatout
- Institute for Environment, Health and Safety, Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium; Department of Molecular Biotechnology, Ghent University, Ghent, Belgium.
| | - An Aerts
- Institute for Environment, Health and Safety, Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| |
Collapse
|
44
|
Parsi M, Desai MH, Desai D, Singhal S, Khandwala PM, Potdar RR. PSMA: a game changer in the diagnosis and treatment of advanced prostate cancer. Med Oncol 2021; 38:89. [PMID: 34181109 DOI: 10.1007/s12032-021-01537-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/15/2021] [Indexed: 12/13/2022]
Abstract
Although management of advanced prostate cancer is evolving, a lot of work remains to be done for patients who have exhausted all options. Molecular targeting of prostate specific membrane antigen (PSMA) is valuable not only for diagnostic but also for therapeutic reasons. PSMA is thus considered to be useful in a theranostic approach. PSMA scans are upcoming diagnostic modalities which detect metastatic lesions that are missed by conventional imaging modalities. PSMA ligand therapy is also an upcoming treatment modality that has been proven to be beneficial with minimal toxicity in patients with advanced prostate cancer that have progressed on prior therapy. In this review article, we summarize the current knowledge regarding PSMA diagnostics and PSMA ligand therapies and discuss their implication in the treatment of advanced prostate cancer.
Collapse
Affiliation(s)
- Meghana Parsi
- Dept of Internal Medicine, Crozer-Chester Medical Center, 1 Medical Center Blvd, Upland, PA, USA.
| | - Milap H Desai
- Drexel University College of Medicine, Philadelphia, PA, USA
| | - Devashish Desai
- Dept of Internal Medicine, Crozer-Chester Medical Center, 1 Medical Center Blvd, Upland, PA, USA
| | - Sachi Singhal
- Dept of Internal Medicine, Crozer-Chester Medical Center, 1 Medical Center Blvd, Upland, PA, USA
| | - Pushti M Khandwala
- Dept of Internal Medicine, Crozer-Chester Medical Center, 1 Medical Center Blvd, Upland, PA, USA
| | - Rashmika R Potdar
- Dept of Hematology Oncology, Crozer-Chester Medical Center, Upland, PA, USA
| |
Collapse
|
45
|
Lankoff A, Czerwińska M, Walczak R, Karczmarczyk U, Tomczyk K, Brzóska K, Fracasso G, Garnuszek P, Mikołajczak R, Kruszewski M. Design and Evaluation of 223Ra-Labeled and Anti-PSMA Targeted NaA Nanozeolites for Prostate Cancer Therapy-Part II. Toxicity, Pharmacokinetics and Biodistribution. Int J Mol Sci 2021; 22:5702. [PMID: 34071854 PMCID: PMC8198605 DOI: 10.3390/ijms22115702] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/24/2021] [Accepted: 05/24/2021] [Indexed: 12/30/2022] Open
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) is a progressive and incurable disease with poor prognosis for patients. Despite introduction of novel therapies, the mortality rate remains high. An attractive alternative for extension of the life of mCRPC patients is PSMA-based targeted radioimmunotherapy. In this paper, we extended our in vitro study of 223Ra-labeled and PSMA-targeted NaA nanozeolites [223RaA-silane-PEG-D2B] by undertaking comprehensive preclinical in vitro and in vivo research. The toxicity of the new compound was evaluated in LNCaP C4-2, DU-145, RWPE-1 and HPrEC prostate cells and in BALB/c mice. The tissue distribution of 133Ba- and 223Ra-labeled conjugates was studied at different time points after injection in BALB/c and LNCaP C4-2 tumor-bearing BALB/c Nude mice. No obvious symptoms of antibody-free and antibody-functionalized nanocarriers cytotoxicity and immunotoxicity was found, while exposure to 223Ra-labeled conjugates resulted in bone marrow fibrosis, decreased the number of WBC and platelets and elevated serum concentrations of ALT and AST enzymes. Biodistribution studies revealed high accumulation of 223Ra-labeled conjugates in the liver, lungs, spleen and bone tissue. Nontargeted and PSMA-targeted radioconjugates exhibited a similar, marginal uptake in tumour lesions. In conclusion, despite the fact that NaA nanozeolites are safe carriers, the intravenous administration of NaA nanozeolite-based radioconjugates is dubious due to its high accumulation in the lungs, liver, spleen and bones.
Collapse
Affiliation(s)
- Anna Lankoff
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (M.C.); (K.B.); (M.K.)
- Department of Medical Biology, Institute of Biology, Jan Kochanowski University, Uniwersytecka 7, 24-406 Kielce, Poland
| | - Malwina Czerwińska
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (M.C.); (K.B.); (M.K.)
| | - Rafał Walczak
- Centre of Radiochemistry and Nuclear Chemistry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland;
| | - Urszula Karczmarczyk
- National Centre for Nuclear Research, Radioisotope Centre POLATOM, Sołtana 7, 05-400 Otwock, Poland; (U.K.); (K.T.); (P.G.); (R.M.)
| | - Kamil Tomczyk
- National Centre for Nuclear Research, Radioisotope Centre POLATOM, Sołtana 7, 05-400 Otwock, Poland; (U.K.); (K.T.); (P.G.); (R.M.)
| | - Kamil Brzóska
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (M.C.); (K.B.); (M.K.)
| | - Giulio Fracasso
- Department of Medicine, University of Verona, 37129 Verona, Italy;
| | - Piotr Garnuszek
- National Centre for Nuclear Research, Radioisotope Centre POLATOM, Sołtana 7, 05-400 Otwock, Poland; (U.K.); (K.T.); (P.G.); (R.M.)
| | - Renata Mikołajczak
- National Centre for Nuclear Research, Radioisotope Centre POLATOM, Sołtana 7, 05-400 Otwock, Poland; (U.K.); (K.T.); (P.G.); (R.M.)
| | - Marcin Kruszewski
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (M.C.); (K.B.); (M.K.)
- Department of Molecular Biology and Translational Research, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland
| |
Collapse
|
46
|
Kovács A. Theoretical Study of Actinide(III)-DOTA Complexes. ACS OMEGA 2021; 6:13321-13330. [PMID: 34056480 PMCID: PMC8158830 DOI: 10.1021/acsomega.1c01292] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 04/20/2021] [Indexed: 06/12/2023]
Abstract
1,4,7,10-Tetraazacyclododecane-N,N',N″,N‴-tetraacetic acid (DOTA) is a prominent chelating ligand used in imaging contrast agents and radiopharmaceuticals. The present study explores the stabilities, structures, and bonding properties of its complexes with trivalent actinides (Ac, U, Np, Pu, Am, Cm, Cf) using density functional theory and relativistic multireference calculations. For reference purposes, the La- and Lu-DOTA complexes are also included. Similar to La3+, the large An3+ ions prefer the TSAP conformer of the ligand. The An-ligand bonding is mainly electrostatic, with minor charge transfer contributions to the An 6d orbitals. For the assessment of the thermodynamic stabilities in aqueous solution, PCM radii to use in conjunction with the SMD solvation model were developed. Basically, the thermodynamic stability of the DOTA complexes increases along the An row but with notable counteracting of spin-orbit coupling.
Collapse
Affiliation(s)
- Attila Kovács
- European Commission Joint
Research Centre, P. O. Box 2340, Karlsruhe D-76125, Germany
| |
Collapse
|
47
|
Hyväkkä A, Virtanen V, Kemppainen J, Grönroos TJ, Minn H, Sundvall M. More Than Meets the Eye: Scientific Rationale behind Molecular Imaging and Therapeutic Targeting of Prostate-Specific Membrane Antigen (PSMA) in Metastatic Prostate Cancer and Beyond. Cancers (Basel) 2021; 13:cancers13092244. [PMID: 34067046 PMCID: PMC8125679 DOI: 10.3390/cancers13092244] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/30/2021] [Accepted: 05/02/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Prostate-specific membrane antigen (PSMA) is a transmembrane protein that is overexpressed in prostate cancer and correlates with the aggressiveness of the disease. PSMA is a promising target for imaging and therapeutics in prostate cancer patients validated in prospective trials. However, the role of PSMA in prostate cancer progression is poorly understood. In this review, we discuss the biology and scientific rationale behind the use of PSMA and other targets in the detection and theranostics of metastatic prostate cancer. Abstract Prostate cancer is the second most common cancer type in men globally. Although the prognosis for localized prostate cancer is good, no curative treatments are available for metastatic disease. Better diagnostic methods could help target therapies and improve the outcome. Prostate-specific membrane antigen (PSMA) is a transmembrane glycoprotein that is overexpressed on malignant prostate tumor cells and correlates with the aggressiveness of the disease. PSMA is a clinically validated target for positron emission tomography (PET) imaging-based diagnostics in prostate cancer, and during recent years several therapeutics have been developed based on PSMA expression and activity. The expression of PSMA in prostate cancer can be very heterogeneous and some metastases are negative for PSMA. Determinants that dictate clinical responses to PSMA-targeting therapeutics are not well known. Moreover, it is not clear how to manipulate PSMA expression for therapeutic purposes and develop rational treatment combinations. A deeper understanding of the biology behind the use of PSMA would help the development of theranostics with radiolabeled compounds and other PSMA-based therapeutic approaches. Along with PSMA several other targets have also been evaluated or are currently under investigation in preclinical or clinical settings in prostate cancer. Here we critically elaborate the biology and scientific rationale behind the use of PSMA and other targets in the detection and therapeutic targeting of metastatic prostate cancer.
Collapse
Affiliation(s)
- Anniina Hyväkkä
- Institute of Biomedicine, Cancer Research Unit, FICAN West Cancer Center Laboratory, University of Turku and Turku University Hospital, FI-20520 Turku, Finland; (A.H.); (V.V.)
| | - Verneri Virtanen
- Institute of Biomedicine, Cancer Research Unit, FICAN West Cancer Center Laboratory, University of Turku and Turku University Hospital, FI-20520 Turku, Finland; (A.H.); (V.V.)
- Turku Doctoral Programme of Molecular Medicine (TuDMM), University of Turku, FI-20520 Turku, Finland
| | - Jukka Kemppainen
- Turku PET Centre, University of Turku, FI-20521 Turku, Finland;
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, FI-20521 Turku, Finland
- Docrates Cancer Center, FI-00180 Helsinki, Finland
| | - Tove J. Grönroos
- Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, FI-20520 Turku, Finland;
| | - Heikki Minn
- Department of Oncology, FICAN West Cancer Center, University of Turku and Turku University Hospital, FI-20521 Turku, Finland;
| | - Maria Sundvall
- Institute of Biomedicine, Cancer Research Unit, FICAN West Cancer Center Laboratory, University of Turku and Turku University Hospital, FI-20520 Turku, Finland; (A.H.); (V.V.)
- Department of Oncology, FICAN West Cancer Center, University of Turku and Turku University Hospital, FI-20521 Turku, Finland;
- Correspondence:
| |
Collapse
|
48
|
Liu G, Tang T, Liu XP, Zhou ZH, Li FJ. Evaluation of the efficiency of peptide receptor radionuclide therapy in patients with metastatic prostate carcinoma: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2021; 100:e25612. [PMID: 33907113 PMCID: PMC8084079 DOI: 10.1097/md.0000000000025612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 04/04/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Metastatic prostate carcinoma has poor prognoses with a median survival period ranging from 2 to 5 years with existing therapeutic challenges. Currently, peptide receptor radionuclide therapy is permitted as a treatment method for metastatic prostate carcinoma patients. Therefore, it is crucial to comprehend the efficiency and safety of peptide receptor radionuclide therapy among this patient population. This study aims to analyse the efficacy of peptide receptor radionuclide therapy when used to treat metastatic prostate carcinoma patients. METHODS This research will perform a methodological search in the following electronic databases to find related randomized controlled trials: Cochrane Library, EMBASE, PubMed, Web of Science, Scopus, China National Knowledge Infrastructure (CNKI), WanFang database, and Chinese BioMedical Literature. All the databases are searched from their inauguration till November 2020. Two independent authors will screen and select literature for review. The two authors will independently utilize the Cochrane Risk of Bias tool to assess the bias risk in studies. This study also plans to conduct subgroup and sensitivity analyses to evaluate the robustness in the results. Statistical analyses will be conducted with the RevMan 5.3 software. RESULTS A high-quality synthesis of existing evidence related to peptide receptor radionuclide therapy in the treatment of metastatic prostate carcinoma will be presented in this study. CONCLUSION Our findings will provide evidence to judge whether peptide receptor radionuclide treatment is efficient for metastatic prostate carcinoma patients. ETHICS AND DISSEMINATION An ethics approval is not required because the data of the present study are primarily obtained from published studies.OSF registration number: December 1, 2020.osf.io/3psx7. (https://osf.io/3psx7/).
Collapse
Affiliation(s)
| | - Tang Tang
- Department of Gynaecology, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai, Guangdong, P.R. China
| | | | | | | |
Collapse
|
49
|
Kgatle MM, Boshomane TMG, Lawal IO, Mokoala KMG, Mokgoro NP, Lourens N, Kairemo K, Zeevaart JR, Vorster M, Sathekge MM. Immune Checkpoints, Inhibitors and Radionuclides in Prostate Cancer: Promising Combinatorial Therapy Approach. Int J Mol Sci 2021; 22:4109. [PMID: 33921181 PMCID: PMC8071559 DOI: 10.3390/ijms22084109] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 01/01/2023] Open
Abstract
Emerging research demonstrates that co-inhibitory immune checkpoints (ICs) remain the most promising immunotherapy targets in various malignancies. Nonetheless, ICIs have offered insignificant clinical benefits in the treatment of advanced prostate cancer (PCa) especially when they are used as monotherapies. Current existing PCa treatment initially offers an improved clinical outcome and overall survival (OS), however, after a while the treatment becomes resistant leading to aggressive and uncontrolled disease associated with increased mortality and morbidity. Concurrent combination of the ICIs with radionuclides therapy that has rapidly emerged as safe and effective targeted approach for treating PCa patients may shift the paradigm of PCa treatment. Here, we provide an overview of the contextual contribution of old and new emerging inhibitory ICs in PCa, preclinical and clinical studies supporting the use of these ICs in treating PCa patients. Furthermore, we will also describe the potential of using a combinatory approach of ICIs and radionuclides therapy in treating PCa patients to enhance efficacy, durable cancer control and OS. The inhibitory ICs considered in this review are cytotoxic T-lymphocyte antigen 4 (CTLA4), programmed cell death 1 (PD1), V-domain immunoglobulin suppressor of T cell activation (VISTA), indoleamine 2,3-dioxygenase (IDO), T cell Immunoglobulin Domain and Mucin Domain 3 (TIM-3), lymphocyte-activation gene 3 (LAG-3), T cell immunoreceptor with Ig and ITIM domains (TIGIT), B7 homolog 3 (B7-H3) and B7-H4.
Collapse
Affiliation(s)
- Mankgopo M. Kgatle
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria 0001, South Africa; (T.M.G.B.); (I.O.L.); (K.M.G.M.); (N.P.M.); (M.V.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa;
| | - Tebatso M. G. Boshomane
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria 0001, South Africa; (T.M.G.B.); (I.O.L.); (K.M.G.M.); (N.P.M.); (M.V.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa;
| | - Ismaheel O. Lawal
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria 0001, South Africa; (T.M.G.B.); (I.O.L.); (K.M.G.M.); (N.P.M.); (M.V.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa;
| | - Kgomotso M. G. Mokoala
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria 0001, South Africa; (T.M.G.B.); (I.O.L.); (K.M.G.M.); (N.P.M.); (M.V.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa;
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria 0001, South Africa
| | - Neo P. Mokgoro
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria 0001, South Africa; (T.M.G.B.); (I.O.L.); (K.M.G.M.); (N.P.M.); (M.V.)
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria 0001, South Africa
| | - Nico Lourens
- Department of Urology, University of Pretoria & Steve Biko Academic Hospital, Pretoria 0001, South Africa;
| | - Kalevo Kairemo
- Departments of Molecular Radiotherapy & Nuclear Medicine, Docrates Cancer Center, 00180 Helsinki, Finland;
- Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jan Rijn Zeevaart
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa;
- Radiochemistry, South African Nuclear Energy Corporation SOC (Necsa), Pelindaba 0001, South Africa
| | - Mariza Vorster
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria 0001, South Africa; (T.M.G.B.); (I.O.L.); (K.M.G.M.); (N.P.M.); (M.V.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa;
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria 0001, South Africa
| | - Mike M. Sathekge
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria 0001, South Africa; (T.M.G.B.); (I.O.L.); (K.M.G.M.); (N.P.M.); (M.V.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa;
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria 0001, South Africa
| |
Collapse
|
50
|
Garnuszek P, Karczmarczyk U, Maurin M, Sikora A, Zaborniak J, Pijarowska-Kruszyna J, Jaroń A, Wyczółkowska M, Wojdowska W, Pawlak D, Lipiński PFJ, Mikołajczak R. PSMA-D4 Radioligand for Targeted Therapy of Prostate Cancer: Synthesis, Characteristics and Preliminary Assessment of Biological Properties. Int J Mol Sci 2021; 22:2731. [PMID: 33800517 PMCID: PMC7962978 DOI: 10.3390/ijms22052731] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/26/2021] [Accepted: 02/26/2021] [Indexed: 12/25/2022] Open
Abstract
A new PSMA ligand (PSMA-D4) containing the Glu-CO-Lys pharmacophore connected with a new linker system (L-Trp-4-Amc) and chelator DOTA was developed for radiolabeling with therapeutic radionuclides. Herein we describe the synthesis, radiolabeling, and preliminary biological evaluation of the novel PSMA-D4 ligand. Synthesized PSMA-D4 was characterized using TOF-ESI-MS, NMR, and HPLC methods. The novel compound was subject to molecular modeling with GCP-II to compare its binding mode to analogous reference compounds. The radiolabeling efficiency of PSMA-D4 with 177Lu, 90Y, 47Sc, and 225Ac was chromatographically tested. In vitro studies were carried out in PSMA-positive LNCaP tumor cells membranes. The ex vivo tissue distribution profile of the radioligands and Cerenkov luminescence imaging (CLI) was studied in LNCaP tumor-bearing mice. PSMA-D4 was synthesized in 24% yield and purity >97%. The radio complexes were obtained with high yields (>97%) and molar activity ranging from 0.11 to 17.2 GBq mcmol-1, depending on the radionuclide. In vitro assays confirmed high specific binding and affinity for all radiocomplexes. Biodistribution and imaging studies revealed high accumulation in LNCaP tumor xenografts and rapid clearance of radiocomplexes from blood and non-target tissues. These render PSMA-D4 a promising ligand for targeted therapy of prostate cancer (PCa) metastases.
Collapse
Affiliation(s)
- Piotr Garnuszek
- National Centre for Nuclear Research, Radioisotope Centre POLATOM, 05-400 Otwock, Poland; (P.G.); (M.M.); (A.S.); (J.P.-K.); (A.J.); (M.W.); (W.W.); (D.P.); (R.M.)
| | - Urszula Karczmarczyk
- National Centre for Nuclear Research, Radioisotope Centre POLATOM, 05-400 Otwock, Poland; (P.G.); (M.M.); (A.S.); (J.P.-K.); (A.J.); (M.W.); (W.W.); (D.P.); (R.M.)
| | - Michał Maurin
- National Centre for Nuclear Research, Radioisotope Centre POLATOM, 05-400 Otwock, Poland; (P.G.); (M.M.); (A.S.); (J.P.-K.); (A.J.); (M.W.); (W.W.); (D.P.); (R.M.)
| | - Arkadiusz Sikora
- National Centre for Nuclear Research, Radioisotope Centre POLATOM, 05-400 Otwock, Poland; (P.G.); (M.M.); (A.S.); (J.P.-K.); (A.J.); (M.W.); (W.W.); (D.P.); (R.M.)
| | | | - Justyna Pijarowska-Kruszyna
- National Centre for Nuclear Research, Radioisotope Centre POLATOM, 05-400 Otwock, Poland; (P.G.); (M.M.); (A.S.); (J.P.-K.); (A.J.); (M.W.); (W.W.); (D.P.); (R.M.)
| | - Antoni Jaroń
- National Centre for Nuclear Research, Radioisotope Centre POLATOM, 05-400 Otwock, Poland; (P.G.); (M.M.); (A.S.); (J.P.-K.); (A.J.); (M.W.); (W.W.); (D.P.); (R.M.)
| | - Monika Wyczółkowska
- National Centre for Nuclear Research, Radioisotope Centre POLATOM, 05-400 Otwock, Poland; (P.G.); (M.M.); (A.S.); (J.P.-K.); (A.J.); (M.W.); (W.W.); (D.P.); (R.M.)
| | - Wioletta Wojdowska
- National Centre for Nuclear Research, Radioisotope Centre POLATOM, 05-400 Otwock, Poland; (P.G.); (M.M.); (A.S.); (J.P.-K.); (A.J.); (M.W.); (W.W.); (D.P.); (R.M.)
| | - Dariusz Pawlak
- National Centre for Nuclear Research, Radioisotope Centre POLATOM, 05-400 Otwock, Poland; (P.G.); (M.M.); (A.S.); (J.P.-K.); (A.J.); (M.W.); (W.W.); (D.P.); (R.M.)
| | - Piotr F. J. Lipiński
- Department of Neuropeptides, Mossakowski Medical Research Center Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Renata Mikołajczak
- National Centre for Nuclear Research, Radioisotope Centre POLATOM, 05-400 Otwock, Poland; (P.G.); (M.M.); (A.S.); (J.P.-K.); (A.J.); (M.W.); (W.W.); (D.P.); (R.M.)
| |
Collapse
|