1
|
Zhou H, Zhong J, Liu Y, Peng S, Yan Q, Wang L, Zhong Y, Hu K. Development of ibuprofen-modified fibroblast activation protein radioligands to improve cancer therapy. Eur J Med Chem 2025; 283:117115. [PMID: 39626520 DOI: 10.1016/j.ejmech.2024.117115] [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: 10/10/2024] [Revised: 11/20/2024] [Accepted: 11/28/2024] [Indexed: 01/03/2025]
Abstract
FAP-targeting radioligands are used in cancer diagnosis and therapy, but their effectiveness is limited by poor tumor uptake and retention. This study aimed to develop new radioligands using an optimized amino acid linker and ibuprofen for better pharmacokinetics. Three novel quinoline-based FAP ligands with an ibuprofen moiety were synthesized and radiolabeled with gallium-68 and lutetium-177. The synthesized FAP ligands FAPI-Ibu1, 2, 3 showed high binding affinity for FAP, with IC50 values of 1.17 ± 0.09, 0.29 ± 0.06, and 0.78 ± 0.12 nM, respectively. 177Lu-labeled FAP ligands showed stability in vitro and demonstrated significant binding to human plasma proteins as well as FAP specificity. PET imaging and biodistribution studies of 68Ga- or 177Lu-labeled FAPI-Ibu1, 2, 3 revealed improved tumor accumulation and retention. Dosimetry calculation showed that [177Lu]Lu-FAPI-Ibu3 delivered a 9.9-fold higher absorbed dose to tumor than [177Lu]Lu-FAPI-04, but only 2.6-fold higher absorbed dose to kidneys leading to 3.8-fold improvement in the tumor-to-kidney absorbed dose ratios. In the endoradiotherapy study, 18.5 MBq of [177Lu]Lu-FAPI-Ibu3 resulted in longer median survival than the equivalent dose of [177Lu]Lu-FAPI-04 (22 vs 16 days). Three ibuprofen-modified FAP radioligands significantly improved tumor uptake, retention, and growth suppression compared to [177Lu]Lu-FAPI-04, with [177Lu]Lu-FAPI-Ibu3 emerging as the most promising candidate for further clinical translational studies.
Collapse
Affiliation(s)
- Hui Zhou
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Jiawei Zhong
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Department of Nuclear Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Yang Liu
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Simin Peng
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qingsong Yan
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lijuan Wang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yuhua Zhong
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Kongzhen Hu
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
2
|
Martin S, Schreck MV, Stemler T, Maus S, Rosar F, Burgard C, Schaefer-Schuler A, Ezziddin S, Bartholomä MD. Development of a homotrimeric PSMA radioligand based on the NOTI chelating platform. EJNMMI Radiopharm Chem 2024; 9:84. [PMID: 39661209 PMCID: PMC11635053 DOI: 10.1186/s41181-024-00314-7] [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] [Accepted: 11/22/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND The NOTI chelating scaffold can readily be derivatized for bioconjugation without impacting its metal complexation/radiolabeling properties making it an attractive building block for the development of multimeric/-valent radiopharmaceuticals. The objective of the study was to further explore the potential of the NOTI chelating platform by preparing and characterizing homotrimeric PSMA radioconjugates in order to identify a suitable candidate for clinical translation. RESULTS Altogether, three PSMA conjugates based on the NOTI-TVA scaffold with different spacer entities between the chelating unit and the Glu-CO-Lys PSMA binding motif were readily prepared by solid phase-peptide chemistry. Cell experiments allowed the identification of the homotrimeric conjugate 9 comprising NaI-Amc spacer with high PSMA binding affinity (IC50 = 5.9 nM) and high PSMA-specific internalization (17.8 ± 2.5%) compared to the clinically used radiotracer [68Ga]Ga-PSMA-11 with a IC50 of 18.5 nM and 5.2 ± 0.2% cell internalization, respectively. All 68Ga-labeled trimeric conjugates showed high metabolic stability in vitro with [68Ga]Ga-9 exhibiting high binding to human serum proteins (> 95%). Small-animal PET imaging revealed a specific tumor uptake of 16.0 ± 1.3% IA g-1 and a kidney uptake of 67.8 ± 8.4% IA g-1 for [68Ga]Ga-9. Clinical PET imaging allowed identification of all lesions detected by [68Ga]Ga-PSMA-11 together with a prolonged blood circulation as well as a significantly lower kidney and higher liver uptake of [68Ga]Ga-9 compared to [68Ga]Ga-PSMA-11. CONCLUSIONS Trimerization of the Glu-CO-Lys binding motif for conjugate 9 resulted in a ~ threefold higher binding affinity and cellular uptake as well as in an altered biodistribution profile compared to the control [68Ga]Ga-PSMA-11 due to its intrinsic high binding to serum proteins. To fully elucidate its biodistribution, future studies in combination with long-lived radionuclides, such as 64Cu, are warranted. Its prolonged biological half-life and favorable tumor-to-kidney ratio make this homotrimeric conjugate also a potential candidate for future radiotherapeutic applications in combination with therapeutic radionuclides such as 67Cu.
Collapse
Affiliation(s)
- Sebastian Martin
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Rue de Bugnon 25A, 1011, Lausanne, Switzerland
| | - Moritz-Valentin Schreck
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, 66421, Homburg, Germany
| | - Tobias Stemler
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, 66421, Homburg, Germany
| | - Stephan Maus
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, 66421, Homburg, Germany
| | - Florian Rosar
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, 66421, Homburg, Germany
| | - Caroline Burgard
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, 66421, Homburg, Germany
| | - Andrea Schaefer-Schuler
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, 66421, Homburg, Germany
| | - Samer Ezziddin
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, 66421, Homburg, Germany
| | - Mark D Bartholomä
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, 66421, Homburg, Germany.
- Department of Nuclear Medicine, Saarland University, Kirrbergerstrasse, 66421, Homburg, Germany.
| |
Collapse
|
3
|
Busslinger SD, Mapanao AK, Kegler K, Bernhardt P, Flühmann F, Fricke J, Zeevaart JR, Köster U, van der Meulen NP, Schibli R, Müller C. Comparison of the tolerability of 161Tb- and 177Lu-labeled somatostatin analogues in the preclinical setting. Eur J Nucl Med Mol Imaging 2024; 51:4049-4061. [PMID: 39046521 PMCID: PMC11527941 DOI: 10.1007/s00259-024-06827-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/30/2024] [Indexed: 07/25/2024]
Abstract
PURPOSE [177Lu]Lu-DOTATATE is an established somatostatin receptor (SSTR) agonist for the treatment of metastasized neuroendocrine neoplasms, while the SSTR antagonist [177Lu]Lu-DOTA-LM3 has only scarcely been employed in clinics. Impressive preclinical data obtained with [161Tb]Tb-DOTA-LM3 in tumor-bearing mice indicated the potential of terbium-161 as an alternative to lutetium-177. The aim of the present study was to compare the tolerability of 161Tb- and 177Lu-based DOTA-LM3 and DOTATATE in immunocompetent mice. METHODS Dosimetry calculations were performed based on biodistribution data of the radiopeptides in immunocompetent mice. Treatment-related effects on blood cell counts were assessed on Days 10, 28 and 56 after application of [161Tb]Tb-DOTA-LM3 or [161Tb]Tb-DOTATATE at 20 MBq per mouse. These radiopeptides were also applied at 100 MBq per mouse and the effects compared to those observed after application of the 177Lu-labeled counterparts. Bone marrow smears, blood plasma parameters and organ histology were assessed at the end of the study. RESULTS The absorbed organ dose was commonly higher for the SSTR antagonist than for the SSTR agonist and for terbium-161 over lutetium-177. Application of a therapeutic activity level of 20 MBq [161Tb]Tb-DOTA-LM3 or [161Tb]Tb-DOTATATE was well tolerated without major hematological changes. The injection of 100 MBq of the 161Tb- and 177Lu-based somatostatin analogues affected the blood cell counts, however. The lymphocytes were 40-50% lower in treated mice compared to the untreated controls on Day 10 irrespective of the radionuclide employed. At the same timepoint, thrombocyte and erythrocyte counts were 30-50% and 6-12% lower, respectively, after administration of the SSTR antagonist (p < 0.05) while changes were less pronounced in mice injected with the SSTR agonist. All blood cell counts were in the normal range on Day 56. Histological analyses revealed minimal abnormalities in the kidneys, liver and spleen of treated mice. No correlation was observed between the organ dose and frequency of the occurrence of abnormalities. CONCLUSION Hematologic changes were more pronounced in mice treated with the SSTR antagonist than in those treated with the SSTR agonist. Despite the increased absorbed dose delivered by terbium-161 over lutetium-177, [161Tb]Tb-DOTA-LM3 and [161Tb]Tb-DOTATATE should be safe at activity levels that are recommended for their respective 177Lu-based analogues.
Collapse
Affiliation(s)
- Sarah D Busslinger
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland
| | - Ana Katrina Mapanao
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland
| | | | - Peter Bernhardt
- Department of Medical Radiation Sciences, Institution of Clinical Science, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 41345, Sweden
- Department of Medical Physics and Biomedical Engineering (MFT), Sahlgrenska University Hospital, Gothenburg, 41345, Sweden
| | - Fabienne Flühmann
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland
| | - Julia Fricke
- Division of Nuclear Medicine, University Hospital Basel, Basel, 4031, Switzerland
| | - Jan Rijn Zeevaart
- Radiochemistry, South African Nuclear Energy Corporation (Necsa), Brits, 0240, South Africa
| | - Ulli Köster
- Institut Laue-Langevin, Grenoble, 38042, France
| | - Nicholas P van der Meulen
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland
- Laboratory of Radiochemistry, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, 8093, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland.
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, 8093, Switzerland.
| |
Collapse
|
4
|
Meher N, Ashley GW, Bobba KN, Wadhwa A, Bidkar AP, Dasari C, Mu C, Sankaranarayanan RA, Serrano JAC, Raveendran A, Bulkley DP, Aggarwal R, Greenland NY, Oskowitz A, Wilson DM, Seo Y, Santi DV, VanBrocklin HF, Flavell RR. Prostate-Specific Membrane Antigen Targeted StarPEG Nanocarrier for Imaging and Therapy of Prostate Cancer. Adv Healthc Mater 2024; 13:e2304618. [PMID: 38700450 PMCID: PMC11281871 DOI: 10.1002/adhm.202304618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 04/29/2024] [Indexed: 05/05/2024]
Abstract
The tumor uptake of large non-targeted nanocarriers primarily occurs through passive extravasation, known as the enhanced permeability and retention (EPR) effect. Prior studies demonstrated improved tumor uptake and retention of 4-arm 40 kDa star polyethylene glycol (StarPEG) polymers for cancer imaging by adding prostate-specific membrane antigen (PSMA) targeting small molecule ligands. To test PSMA-targeted delivery and therapeutic efficacy, StarPEG nanodrugs with/without three copies of PSMA-targeting ligands, ACUPA, are designed and synthesized. For single-photon emission computed tomography (SPECT) imaging and therapy, each nanocarrier is labeled with 177Lu using DOTA radiometal chelator. The radiolabeled nanodrugs, [177Lu]PEG-(DOTA)1 and [177Lu]PEG-(DOTA)1(ACUPA)3, are evaluated in vitro and in vivo using PSMA+ PC3-Pip and/or PSMA- PC3-Flu cell lines, subcutaneous xenografts and disseminated metastatic models. The nanocarriers are efficiently radiolabeled with 177Lu with molar activities 10.8-15.8 MBq/nmol. Besides excellent in vitro PSMA binding affinity (kD = 51.7 nM), the targeted nanocarrier, [177Lu]PEG-(DOTA)1(ACUPA)3, demonstrated excellent in vivo SPECT imaging contrast with 21.3% ID/g PC3-Pip tumors uptake at 192 h. Single doses of 18.5 MBq [177Lu]PEG-(DOTA)1(ACUPA)3 showed complete resolution of the PC3-Pip xenografts observed up to 138 days. Along with PSMA-targeted excellent imaging contrast, these results demonstrated high treatment efficacy of [177Lu]PEG-(DOTA)1(ACUPA)3 for prostate cancer, with potential for clinical translation.
Collapse
Affiliation(s)
- Niranjan Meher
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, United States
- National Institute of Pharmaceutical Education and Research, Raebareli, Lucknow, UP 226002, India
| | | | - Kondapa Naidu Bobba
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, United States
| | - Anju Wadhwa
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, United States
| | - Anil P. Bidkar
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, United States
| | - Chandrashekhar Dasari
- Division of Vascular and Endovascular Surgery, University of California, San Francisco, CA 94143-0957, United States
| | - Changhua Mu
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, United States
| | - Ramya Ambur Sankaranarayanan
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, United States
| | - Juan A. Camara Serrano
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143-0981, United States
| | - Athira Raveendran
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, United States
| | - David P. Bulkley
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, United States
| | - Rahul Aggarwal
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143-0981, United States
| | - Nancy Y. Greenland
- Department of Pathology, University of California, San Francisco, CA 94143, United States
| | - Adam Oskowitz
- Division of Vascular and Endovascular Surgery, University of California, San Francisco, CA 94143-0957, United States
| | - David M. Wilson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, United States
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143-0981, United States
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, United States
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143-0981, United States
| | | | - Henry F. VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, United States
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143-0981, United States
| | - Robert R. Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, United States
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143-0981, United States
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158-2517, United States
| |
Collapse
|
5
|
Pun MD, Gallazzi F, Ho KV, Watkinson L, Carmack TL, Iweha E, Li L, Anderson CJ. Albumin-Binding Lutetium-177-Labeled LLP2A Derivatives as Theranostics for Melanoma. Mol Pharm 2024; 21:2960-2969. [PMID: 38680059 DOI: 10.1021/acs.molpharmaceut.4c00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Very late antigen-4 (VLA-4) is a transmembrane integrin protein that is highly expressed in aggressive forms of metastatic melanoma. A small-molecule peptidomimetic, LLP2A, was found to have a low pM affinity binding to VLA-4. Because LLP2A itself does not inhibit cancer cell proliferation and survival, it is an ideal candidate for the imaging and delivery of therapeutic payloads. An analog of [177Lu]Lu-labeled-LLP2A was previously investigated as a therapeutic agent in melanoma tumor-bearing mice, resulting in only a modest improvement in tumor growth inhibition, likely due to rapid clearance of the agent from the tumor. To improve the pharmacokinetic profile, DOTAGA-PEG4-LLP2A with a 4-(p-iodophenyl)butyric acid (pIBA) albumin binding moiety was synthesized. We demonstrate the feasibility of this albumin binding strategy by comparing in vitro cell binding assays and in vivo biodistribution performance of [177Lu]Lu-DOTAGA-PEG4-LLP2A ([177Lu]Lu-1) to the albumin binding [177Lu]Lu-DOTAGA-pIBA-PEG4-LLP2A ([177Lu]Lu-2). In vitro cell binding assay results for [177Lu]Lu-1 and [177Lu]Lu-2 showed Kd values of 0.40 ± 0.07 and 1.75 ± 0.40 nM, with similar Bmax values of 200 ± 6 and 315 ± 15 fmol/mg, respectively. In vivo biodistribution data for both tracers exhibited specific uptake in the tumor, spleen, thymus, and bone due to endogenous expression of VLA-4. Compound [177Lu]Lu-2 exhibited a much longer blood circulation time compared to [177Lu]Lu-1. The tumor uptake for [177Lu]Lu-1 was highest at 1 h (∼15%ID/g) and that for [177Lu]Lu-2 was highest at 4 h (∼23%ID/g). Significant clearance of [177Lu]Lu-1 from the tumor occurs at 24 h (<5%ID/g) while[177Lu]Lu-2 is retained for greater than 96 h (∼10%ID/g). An efficacy study showed that melanoma tumor-bearing mice receiving compound [177Lu]Lu-2 given in two fractions (2 × 14.8 MBq, 14 days apart) had a greater median survival time than mice administered a single 29.6 MBq dose of compound [177Lu]Lu-1, while a single 29.6 MBq dose of [177Lu]Lu-2 imparted hematopoietic toxicity. The in vitro and in vivo data show addition of pIBA to [177Lu]Lu-DOTAGA-PEG4-LLP2A slows blood clearance for a higher tumor uptake, and there is potential of [177Lu]Lu-2 as a theranostic in fractionated administered doses.
Collapse
Affiliation(s)
- Michael D Pun
- Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Fabio Gallazzi
- Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
- Molecular Interactions Core, University of Missouri, Columbia, Missouri 65211, United States
| | - Khanh-Van Ho
- Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Lisa Watkinson
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
- Research Division, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri 65211, United States
- University of Missouri Research Reactor Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Terry L Carmack
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
- Research Division, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri 65211, United States
- University of Missouri Research Reactor Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Ejike Iweha
- Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Longbo Li
- Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Carolyn J Anderson
- Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
- Department of Radiology, University of Missouri, Columbia, Missouri 65212, United States
- Ellis Fischel Cancer Center, University of Missouri, Columbia, Missouri 65212, United States
| |
Collapse
|
6
|
Kristiansson A, Vilhelmsson Timmermand O, Altai M, Strand SE, Åkerström B, Örbom A. Hematological and renal toxicity in mice after three cycles of high activity [ 177Lu]Lu-PSMA-617 with or without human α 1-microglobulin. Sci Rep 2024; 14:10787. [PMID: 38734765 PMCID: PMC11088679 DOI: 10.1038/s41598-024-61370-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/06/2024] [Indexed: 05/13/2024] Open
Abstract
Radioligand therapy with [177Lu]Lu-PSMA-617 can be used to prolong life and reduce tumor burden in terminally ill castration resistant prostate cancer patients. Still, accumulation in healthy tissue limits the activity that can be administered. Therefore, fractionated therapy is used to lower toxicity. However, there might be a need to reduce toxicity even further with e.g. radioprotectors. The aim of this study was to (i). establish a preclinical mouse model with fractionated high activity therapy of three consecutive doses of 200 MBq [177Lu]Lu-PSMA-617 in which we aimed to (ii). achieve measurable hematotoxicity and nephrotoxicity and to (iii). analyze the potential protective effect of co-injecting recombinant α1-microglobulin (rA1M), a human antioxidant previously shown to have radioprotective effects. In both groups, three cycles resulted in increased albuminuria for each cycle, with large individual variation. Another marker of kidney injury, serum blood urea nitrogen (BUN), was only significantly increased compared to control animals after the third cycle. The number of white and red blood cells decreased significantly and did not reach the levels of control animals during the experiment. rA1M did reduce absorbed dose to kidney but did not show significant protection here, but future studies are warranted due to the recent clinical studies showing a significant renoprotective effect in patients.
Collapse
Affiliation(s)
- Amanda Kristiansson
- Department of Clinical Sciences Lund, Section for Oncology, Lund University, Barngatan 4, 222 42, Lund, Sweden
- Department of Clinical Sciences Lund, Section for Pediatrics, Lund University, Lund, Sweden
- Department of Neonatology, Skåne University Hospital, Lund, Sweden
| | - Oskar Vilhelmsson Timmermand
- Department of Clinical Sciences Lund, Section for Oncology, Lund University, Barngatan 4, 222 42, Lund, Sweden
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Mohamed Altai
- Department of Clinical Sciences Lund, Section for Oncology, Lund University, Barngatan 4, 222 42, Lund, Sweden
| | - Sven-Erik Strand
- Department of Clinical Sciences Lund, Section for Oncology, Lund University, Barngatan 4, 222 42, Lund, Sweden
- Department of Clinical Sciences Lund, Section for Medical Radiation Physics, Lund University, Lund, Sweden
| | - Bo Åkerström
- Department of Clinical Sciences Lund, Section for Infection Medicine, Lund University, Lund, Sweden
| | - Anders Örbom
- Department of Clinical Sciences Lund, Section for Oncology, Lund University, Barngatan 4, 222 42, Lund, Sweden.
| |
Collapse
|
7
|
Mattana F, Muraglia L, Barone A, Colandrea M, Saker Diffalah Y, Provera S, Cascio AS, Omodeo Salè E, Ceci F. Prostate-Specific Membrane Antigen-Targeted Therapy in Prostate Cancer: History, Combination Therapies, Trials, and Future Perspective. Cancers (Basel) 2024; 16:1643. [PMID: 38730595 PMCID: PMC11083597 DOI: 10.3390/cancers16091643] [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: 03/25/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
In the last decades, the development of PET/CT radiopharmaceuticals, targeting the Prostate-Specific Membrane Antigen (PSMA), changed the management of prostate cancer (PCa) patients thanks to its higher diagnostic accuracy in comparison with conventional imaging both in staging and in recurrence. Alongside molecular imaging, PSMA was studied as a therapeutic agent targeted with various isotopes. In 2021, results from the VISION trial led to the Food and Drug Administration (FDA) approval of [177Lu]Lu-PSMA-617 as a novel therapy for metastatic castration-resistant prostate cancer (mCRPC) and set the basis for a radical change in the future perspectives of PCa treatment and the history of Nuclear Medicine. Despite these promising results, primary resistance in patients treated with single-agent [177Lu]Lu-PSMA-617 remains a real issue. Emerging trials are investigating the use of [177Lu]Lu-PSMA-617 in combination with other PCa therapies in order to cover the multiple oncologic resistance pathways and to overcome tumor heterogeneity. In this review, our aim is to retrace the history of PSMA-targeted therapy from the first preclinical studies to its future applications in PCa.
Collapse
Affiliation(s)
- Francesco Mattana
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (A.B.); (M.C.); (A.S.C.); (F.C.)
| | - Lorenzo Muraglia
- Division of Nuclear Medicine, Humanitas IRCCS, 20141 Milan, Italy;
| | - Antonio Barone
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (A.B.); (M.C.); (A.S.C.); (F.C.)
| | - Marzia Colandrea
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (A.B.); (M.C.); (A.S.C.); (F.C.)
| | - Yasmina Saker Diffalah
- Division of Nuclear Medicine, Hospital Clínico Universitario Lozano Blesa, 50009 Zaragoza, Spain;
| | - Silvia Provera
- Division of Pharmacy, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (S.P.); (E.O.S.)
| | - Alfio Severino Cascio
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (A.B.); (M.C.); (A.S.C.); (F.C.)
| | - Emanuela Omodeo Salè
- Division of Pharmacy, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (S.P.); (E.O.S.)
| | - Francesco Ceci
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (A.B.); (M.C.); (A.S.C.); (F.C.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| |
Collapse
|
8
|
Song Y, Zou J, Castellanos EA, Matsuura N, Ronald JA, Shuhendler A, Weber WA, Gilad AA, Müller C, Witney TH, Chen X. Theranostics - a sure cure for cancer after 100 years? Theranostics 2024; 14:2464-2488. [PMID: 38646648 PMCID: PMC11024861 DOI: 10.7150/thno.96675] [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: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/23/2024] Open
Abstract
Cancer has remained a formidable challenge in medicine and has claimed an enormous number of lives worldwide. Theranostics, combining diagnostic methods with personalized therapeutic approaches, shows huge potential to advance the battle against cancer. This review aims to provide an overview of theranostics in oncology: exploring its history, current advances, challenges, and prospects. We present the fundamental evolution of theranostics from radiotherapeutics, cellular therapeutics, and nanotherapeutics, showcasing critical milestones in the last decade. From the early concept of targeted drug delivery to the emergence of personalized medicine, theranostics has benefited from advances in imaging technologies, molecular biology, and nanomedicine. Furthermore, we emphasize pertinent illustrations showcasing that revolutionary strategies in cancer management enhance diagnostic accuracy and provide targeted therapies customized for individual patients, thereby facilitating the implementation of personalized medicine. Finally, we describe future perspectives on current challenges, emerging topics, and advances in the field.
Collapse
Affiliation(s)
- Yangmeihui Song
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, 81675, Germany
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 43000, China
| | - Jianhua Zou
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | | | - Naomi Matsuura
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Department of Materials Science & Engineering, University of Toronto, Toronto, ON, Canada
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - John A. Ronald
- Imaging Laboratories, Department of Medical Biophysics, Robarts Research Institute, University of Western Ontario, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
| | - Adam Shuhendler
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Wolfgang A Weber
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, 81675, Germany
| | - Assaf A. Gilad
- Department of Chemical Engineering and Materials Sciences, Michigan State University, East Lansing, MI, USA
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, USA
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Timothy H. Witney
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
9
|
de Roode KE, Joosten L, Behe M. Towards the Magic Radioactive Bullet: Improving Targeted Radionuclide Therapy by Reducing the Renal Retention of Radioligands. Pharmaceuticals (Basel) 2024; 17:256. [PMID: 38399470 PMCID: PMC10892921 DOI: 10.3390/ph17020256] [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: 06/23/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Targeted radionuclide therapy (TRT) is an emerging field and has the potential to become a major pillar in effective cancer treatment. Several pharmaceuticals are already in routine use for treating cancer, and there is still a high potential for new compounds for this application. But, a major issue for many radiolabeled low-to-moderate-molecular-weight molecules is their clearance via the kidneys and their subsequent reuptake. High renal accumulation of radioactive compounds may lead to nephrotoxicity, and therefore, the kidneys are often the dose-limiting organs in TRT with these radioligands. Over the years, different strategies have been developed aiming for reduced kidney retention and enhanced therapeutic efficacy of radioligands. In this review, we will give an overview of the efforts and achievements of the used strategies, with focus on the therapeutic potential of low-to-moderate-molecular-weight molecules. Among the strategies discussed here is coadministration of compounds that compete for binding to the endocytic receptors in the proximal tubuli. In addition, the influence of altering the molecular design of radiolabeled ligands on pharmacokinetics is discussed, which includes changes in their physicochemical properties and implementation of cleavable linkers or albumin-binding moieties. Furthermore, we discuss the influence of chelator and radionuclide choice on reabsorption of radioligands by the kidneys.
Collapse
Affiliation(s)
- Kim E. de Roode
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands;
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | - Lieke Joosten
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands;
| | - Martin Behe
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, 5232 Villigen, Switzerland
| |
Collapse
|
10
|
Sangwan K, Sharma V, Goyal PK. Pharmacological Profile of Novel Anti-cancer Drugs Approved by USFDA in 2022: A Review. Curr Mol Med 2024; 24:734-750. [PMID: 37350009 DOI: 10.2174/1566524023666230622151034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND For any drug molecule, it is mandatory to pass the drug approval process of the concerned regulatory authority, before being marketed. The Food and Drug Administration (FDA), throughout the year, approves several new drugs for safety and efficacy. In addition to new drug approvals, FDA also works on improving access to generic drugs, aimed to lower the cost of drugs for patients and improve access to treatments. In the year 2022 twelve new drug therapies were approved for managing varying cancers. METHODS This manuscript is focused to describe the pharmacological aspects including therapeutic uses, mechanisms of actions, pharmacokinetics, adverse effects, doses, indication for special cases, contraindications, etc., of novel FDA-approved anticancer drug therapies in the year 2022. RESULT FDA has approved about 29% (11 out of 37) novel drug therapies for varying types of cancers such as lung cancer, breast cancer, prostate cancer, melanoma, leukemia, etc. The Center for Drug Evaluation and Research CDER has reported that 90% of these anticancer drugs (e.g. Adagrasib, Futibatinib, Mirvetuximabsoravtansinegynx, Mosunetuzumab-axb, Nivolumab and relatlimab-rmbw, Olutasidenib, Pacritinib, Tebentafusp-tebn, Teclistamab-cqyv, and Tremelimumab-actl) as orphan drugs and recommended to treat rare or uncommon cancers such as non-small cell lung cancer, metastatic intrahepatic cholangio-carcinoma, epithelial ovarian cancer, follicular lymphoma, metastatic melanoma, metastatic uveal melanoma, etc. CDER has identified six anticancer drugs (e.g. Lutetium (177Lu)vipivotidetetraxetan, Mirvetuximabsoravtansine- gynx, Mosunetuzumab-axb, Nivolumab and relatlimab-rmbw, Tebentafusp-tebn, Teclistamab-cqyv) as first-in-class drugs i.e. drugs having different mechanisms of action from the already existing ones. The newly approved anticancer drugs shall provide more efficient treatment options for cancer patients. Three FDA-approved anticancer drugs in the year 2023 are also briefly described in the manuscript. CONCLUSION This manuscript, describing the pharmacological aspects of eleven anticancer novel drug therapies approved by the FDA, shall serve as a helpful document for cancer patients, concerned academicians, researchers, and clinicians, especially oncologists.
Collapse
Affiliation(s)
- Kavita Sangwan
- Department of Pharmacy, Panipat Institute of Engineering and Technology (PIET), Samalkha, Panipat, 132102, Haryana, India
| | - Vipasha Sharma
- Department of Pharmacy, Panipat Institute of Engineering and Technology (PIET), Samalkha, Panipat, 132102, Haryana, India
| | - Parveen Kumar Goyal
- Department of Pharmacy, Panipat Institute of Engineering and Technology (PIET), Samalkha, Panipat, 132102, Haryana, India
| |
Collapse
|
11
|
Qin S, Yang Y, Zhang J, Yin Y, Liu W, Zhang H, Fan X, Yang M, Yu F. Effective Treatment of SSTR2-Positive Small Cell Lung Cancer Using 211At-Containing Targeted α-Particle Therapy Agent Which Promotes Endogenous Antitumor Immune Response. Mol Pharm 2023; 20:5543-5553. [PMID: 37788300 PMCID: PMC10630944 DOI: 10.1021/acs.molpharmaceut.3c00427] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/15/2023] [Accepted: 09/15/2023] [Indexed: 10/05/2023]
Abstract
Small cell lung cancer (SCLC) is a neuroendocrine tumor with a high degree of malignancy. Due to limited treatment options, patients with SCLC have a poor prognosis. We have found, however, that intravenously administered octreotide (Oct) armed with astatine-211 ([211At]SAB-Oct) is effective against a somatostatin receptor 2 (SSTR2)-positive SCLC tumor in SCLC tumor-bearing BALB/c nude mice. In biodistribution analysis, [211At]SAB-Oct achieved the highest concentration in the SCLC tumors up to 3 h after injection as time proceeded. A single intravenous injection of [211At]SAB-Oct (370 kBq) was sufficient to suppress SSTR2-positive SCLC tumor growth in treated mice by inducing DNA double-strand breaks. Additionally, a multitreatment course (370 kBq followed by twice doses of 370 kBq for a total of 1110 kBq) inhibited the growth of the tumor compared to the untreated control group without significant off-target toxicity. Surprisingly, we found that [211At]SAB-Oct could up-regulate the expressions of calreticulin and major histocompatibility complex I (MHC-I) on the tumor cell membrane surface, suggesting that α-particle internal irradiation may activate an endogenous antitumor immune response through the regulation of immune cells in the tumor microenvironment, which could synergically enhance the efficacy of immunotherapy. We conclude that [211At]SAB-Oct is a potential new therapeutic option for SSTR2-positive SCLC.
Collapse
Affiliation(s)
- Shanshan Qin
- Department
of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, People’s Republic of China
- Institute
of Nuclear Medicine, Tongji University School
of Medicine, No. 301
Yan-chang-zhong Road, Shanghai 200072, People’s Republic
of China
| | - Yuanyou Yang
- Key
Laboratory of Radiation Physics and Technology, Ministry of Education,
Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, People’s
Republic of China
| | - Jiajia Zhang
- Department
of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, People’s Republic of China
- Institute
of Nuclear Medicine, Tongji University School
of Medicine, No. 301
Yan-chang-zhong Road, Shanghai 200072, People’s Republic
of China
| | - Yuzhen Yin
- Department
of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, People’s Republic of China
- Institute
of Nuclear Medicine, Tongji University School
of Medicine, No. 301
Yan-chang-zhong Road, Shanghai 200072, People’s Republic
of China
| | - Weihao Liu
- Key
Laboratory of Radiation Physics and Technology, Ministry of Education,
Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, People’s
Republic of China
| | - Han Zhang
- Department
of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, People’s Republic of China
- Institute
of Nuclear Medicine, Tongji University School
of Medicine, No. 301
Yan-chang-zhong Road, Shanghai 200072, People’s Republic
of China
| | - Xin Fan
- Department
of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, People’s Republic of China
- Institute
of Nuclear Medicine, Tongji University School
of Medicine, No. 301
Yan-chang-zhong Road, Shanghai 200072, People’s Republic
of China
| | - Mengdie Yang
- Department
of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, People’s Republic of China
- Institute
of Nuclear Medicine, Tongji University School
of Medicine, No. 301
Yan-chang-zhong Road, Shanghai 200072, People’s Republic
of China
| | - Fei Yu
- Department
of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, People’s Republic of China
- Institute
of Nuclear Medicine, Tongji University School
of Medicine, No. 301
Yan-chang-zhong Road, Shanghai 200072, People’s Republic
of China
| |
Collapse
|
12
|
Tschan VJ, Busslinger SD, Bernhardt P, Grundler PV, Zeevaart JR, Köster U, van der Meulen NP, Schibli R, Müller C. Albumin-Binding and Conventional PSMA Ligands in Combination with 161Tb: Biodistribution, Dosimetry, and Preclinical Therapy. J Nucl Med 2023; 64:1625-1631. [PMID: 37442604 DOI: 10.2967/jnumed.123.265524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 05/31/2023] [Indexed: 07/15/2023] Open
Abstract
The favorable decay characteristics of 161Tb attracted the interest of clinicians in using this novel radionuclide for radioligand therapy (RLT). 161Tb decays with a similar half-life to 177Lu, but beyond the emission of β--particles and γ-rays, 161Tb also emits conversion and Auger electrons, which may be particularly effective to eliminate micrometastases. The aim of this study was to compare the dosimetry and therapeutic efficacy of 161Tb and 177Lu in tumor-bearing mice using SibuDAB and PSMA-I&T, which differ in their blood residence time and tumor uptake. Methods: [161Tb]Tb-SibuDAB and [161Tb]Tb-PSMA-I&T were evaluated in vitro and investigated in biodistribution, imaging, and therapy studies using PC-3 PIP tumor-bearing mice. The 177Lu-labeled counterparts served for dose calculations and comparison of therapeutic efficacy. The tolerability of RLT in mice was monitored on the basis of body mass, blood plasma parameters, blood cell counts, and the histology of relevant organs and tissues. Results: The prostate-specific membrane antigen (PSMA)-targeting radioligands, irrespective of whether labeled with 161Tb or 177Lu, showed similar in vitro data and comparable tissue distribution profiles. As a result of the albumin-binding properties, [161Tb]Tb/[177Lu]Lu-SibuDAB had an enhanced blood residence time and higher tumor uptake (62%-69% injected activity per gram at 24 h after injection) than [161Tb]Tb/[177Lu]Lu-PSMA-I&T (30%-35% injected activity per gram at 24 h after injection). [161Tb]Tb-SibuDAB inhibited tumor growth more effectively than [161Tb]Tb-PSMA-I&T, as can be ascribed to its 4-fold increased absorbed tumor dose. At any of the applied activities, the 161Tb-based radioligands were therapeutically more effective than their 177Lu-labeled counterparts, as agreed with the approximately 40% increased tumor dose of 161Tb compared with that of 177Lu. Under the given experimental conditions, no obvious adverse events were observed. Conclusion: The data of this study indicate the promising potential of 161Tb in combination with SibuDAB for RLT of prostate cancer. Future clinical studies using 161Tb-based RLT will shed light on a potential clinical benefit of 161Tb over 177Lu.
Collapse
Affiliation(s)
- Viviane J Tschan
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Sarah D Busslinger
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Peter Bernhardt
- Department of Radiation Physics, Institution of Clinical Science, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pascal V Grundler
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Jan Rijn Zeevaart
- Radiochemistry, South African Nuclear Energy Corporation (Necsa), Brits, South Africa
| | | | - Nicholas P van der Meulen
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland
- Laboratory of Radiochemistry, Paul Scherrer Institute, Villigen-PSI, Switzerland; and
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland;
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Alati S, Singh R, Pomper MG, Rowe SP, Banerjee SR. Preclinical Development in Radiopharmaceutical Therapy for Prostate Cancer. Semin Nucl Med 2023; 53:663-686. [PMID: 37468417 DOI: 10.1053/j.semnuclmed.2023.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/21/2023]
Abstract
Prostate cancer is a leading cause of cancer death in men worldwide. Among the various treatment options, radiopharmaceutical therapy has shown notable success in metastatic, castration-resistant disease. Radiopharmaceutical therapy is a systemic approach that delivers cytotoxic radiation doses precisely to the malignant tumors and/or tumor microenvironment. Therapeutic radiopharmaceuticals are composed of a therapeutic radionuclide and a high-affinity, tumor-targeting carrier molecule. Therapeutic radionuclides used in preclinical prostate cancer studies are primarily α-, β--, or Auger-electron-emitting radiometals or radiohalogens. Monoclonal antibodies, antibody-derived fragments, peptides, and small molecules are frequently used as tumor-targeting molecules. Over the years, several important membrane-associated proteases and receptors have been identified, validated, and subsequently used for preclinical radiotherapeutic development for prostate cancer. Prostate-specific membrane antigen (PSMA) is the most well-studied prostate cancer-associated protease in preclinical literature. PSMA-targeting radiotherapeutic agents are being investigated using high-affinity antibody- and small-molecule-based agents for safety and efficacy. Early generations of such agents were developed simply by replacing radionuclides of the imaging agents with therapeutic ones. Later, extensive structure-activity relationship studies were conducted to address the safety and efficacy issues obtained from initial patient data. Recent regulatory approval of the 177Lu-labeled low-molecular-weight agent, 177Lu-PSMA-617, is a significant accomplishment. Current preclinical experiments are focused on the structural modification of 177Lu-PSMA-617 and relevant investigational agents to increase tumor targeting and reduce off-target binding and toxicity in healthy organs. While lutetium-177 (177Lu) remains the most widely used radionuclide, radiolabeled analogs with iodine-131 (128I), yttrium-90 (89Y), copper-67 (67Cu), and terbium-161 (161Tb) have been evaluated as potential alternatives in recent years. In addition, agents carrying the α-particle-emitting radiohalogen, astatine-211 (211At), or radiometals, actinium-225 (225Ac), lead-212 (212Pb), radium-223 (223Ra), and thorium-227 (227Th), have been increasingly investigated in preclinical research. Besides PSMA-based radiotherapeutics, other prominent prostate cancer-related proteases, for example, human kallikrein peptidases (HK2 and HK3), have been explored using monoclonal-antibody-(mAb)-based targeting platforms. Several promising mAbs targeting receptors overexpressed on the different stages of prostate cancer have also been developed for radiopharmaceutical therapy, for example, Delta-like ligand 3 (DLL-3), CD46, and CUB domain-containing protein 1 (CDCP1). Progress is also being made using peptide-based targeting platforms for the gastrin-releasing peptide receptor (GRPR), a well-established membrane-associated receptor expressed in localized and metastatic prostate cancers. Furthermore, mechanism-driven combination therapies appear to be a burgeoning area in the context of preclinical prostate cancer radiotherapeutics. Here, we review the current developments related to the preclinical radiopharmaceutical therapy of prostate cancer. These are summarized in two major topics: (1) therapeutic radionuclides and (2) tumor-targeting approaches using monoclonal antibodies, small molecules, and peptides.
Collapse
Affiliation(s)
- Suresh Alati
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Rajan Singh
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Steven P Rowe
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Sangeeta Ray Banerjee
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD.
| |
Collapse
|
14
|
Boinapally S, Alati S, Jiang Z, Yan Y, Lisok A, Singh R, Lofland G, Minn I, Hobbs RF, Pomper MG, Banerjee SR. Preclinical Evaluation of a New Series of Albumin-Binding 177Lu-Labeled PSMA-Based Low-Molecular-Weight Radiotherapeutics. Molecules 2023; 28:6158. [PMID: 37630410 PMCID: PMC10459686 DOI: 10.3390/molecules28166158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/12/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA)-based low-molecular-weight agents using beta(β)-particle-emitting radiopharmaceuticals is a new treatment paradigm for patients with metastatic castration-resistant prostate cancer. Although results have been encouraging, there is a need to improve the tumor residence time of current PSMA-based radiotherapeutics. Albumin-binding moieties have been used strategically to enhance the tumor uptake and retention of existing PSMA-based investigational agents. Previously, we developed a series of PSMA-based, β-particle-emitting, low-molecular-weight compounds. From this series, 177Lu-L1 was selected as the lead agent because of its reduced off-target radiotoxicity in preclinical studies. The ligand L1 contains a PSMA-targeting Lys-Glu urea moiety with an N-bromobenzyl substituent in the ε-amino group of Lys. Here, we structurally modified 177Lu-L1 to improve tumor targeting using two known albumin-binding moieties, 4-(p-iodophenyl) butyric acid moiety (IPBA) and ibuprofen (IBU), and evaluated the effects of linker length and composition. Six structurally related PSMA-targeting ligands (Alb-L1-Alb-L6) were synthesized based on the structure of 177Lu-L1. The ligands were assessed for in vitro binding affinity and were radiolabeled with 177Lu following standard protocols. All 177Lu-labeled analogs were studied in cell uptake and selected cell efficacy studies. In vivo pharmacokinetics were investigated by conducting tissue biodistribution studies for 177Lu-Alb-L2-177Lu-Alb-L6 (2 h, 24 h, 72 h, and 192 h) in male NSG mice bearing human PSMA+ PC3 PIP and PSMA- PC3 flu xenografts. Preliminary therapeutic ratios of the agents were estimated from the area under the curve (AUC0-192h) of the tumors, blood, and kidney uptake values. Compounds were obtained in >98% radiochemical yields and >99% purity. PSMA inhibition constants (Kis) of the ligands were in the ≤10 nM range. The long-linker-based agents, 177Lu-Alb-L4 and 177Lu-Alb-L5, displayed significantly higher tumor uptake and retention (p < 0.001) than the short-linker-bearing 177Lu-Alb-L2 and 177Lu-Alb-L3 and a long polyethylene glycol (PEG) linker-bearing agent, 177Lu-Alb-L6. The area under the curve (AUC0-192h) of the PSMA+ PC3 PIP tumor uptake of 177Lu-Alb-L4 and 177Lu-Alb-L5 were >4-fold higher than 177Lu-Alb-L2, 177Lu-Alb-L3, and 177Lu-Alb-L6, respectively. Also, the PSMA+ PIP tumor uptake (AUC0-192h) of 177Lu-Alb-L2 and 177Lu-Alb-L3 was ~1.5-fold higher than 177Lu-Alb-L6. However, the lowest blood AUC0-192h and kidney AUC0-192h were associated with 177Lu-Alb-L6 from the series. Consequently, 177Lu-Alb-L6 displayed the highest ratios of AUC(tumor)-to-AUC(blood) and AUC(tumor)-to-AUC(kidney) values from the series. Among the other agents, 177Lu-Alb-L4 demonstrated a nearly similar ratio of AUC(tumor)-to-AUC(blood) as 177Lu-Alb-L6. The tumor-to-blood ratio was the dose-limiting therapeutic ratio for all of the compounds. Conclusions: 177Lu-Alb-L4 and 177Lu-Alb-L6 showed high tumor uptake in PSMA+ tumors and tumor-to-blood ratios. The data suggest that linker length and composition can be modulated to generate an optimized therapeutic agent.
Collapse
Affiliation(s)
- Srikanth Boinapally
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Suresh Alati
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Zirui Jiang
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Yu Yan
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Alla Lisok
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Rajan Singh
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Gabriela Lofland
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Il Minn
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Robert F. Hobbs
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Martin G. Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Sangeeta Ray Banerjee
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA
| |
Collapse
|
15
|
Deshayes E, Fersing C, Thibault C, Roumiguie M, Pourquier P, Houédé N. Innovation in Radionuclide Therapy for the Treatment of Prostate Cancers: Radiochemical Perspective and Recent Therapeutic Practices. Cancers (Basel) 2023; 15:3133. [PMID: 37370743 DOI: 10.3390/cancers15123133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/31/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Prostate cancer represents the second cause of death by cancer in males in western countries. While early-stage diseases are accessible to surgery and/or external radiotherapy, advanced metastatic prostate cancers are primarily treated with androgen deprivation therapy, to which new generation androgen receptor antagonists or taxane-based chemotherapies are added in the case of tumor relapse. Nevertheless, patients become invariably resistant to castration with a median survival that rarely exceeds 3 years. This fostered the search for alternative strategies, independent of the androgen receptor signaling pathway. In this line, radionuclide therapies may represent an interesting option as they could target either the microenvironment of sclerotic bone metastases with the use of radiopharmaceuticals containing samarium-153, strontium-89 or radium-223 or tumor cells expressing the prostate-specific membrane antigen (PSMA), a protein found at the surface of prostate cancer cells. This review gives highlights the chemical properties of radioligands targeting prostate cancer cells and recapitulates the clinical trials evaluating the efficacy of radionuclide therapies, alone or in combination with other approved treatments, in patients with castration-resistant prostate tumors. It discusses some of the encouraging results obtained, especially the benefit on overall survival that was reported with [177Lu]-PSMA-617. It also addresses the specific requirements for the use of this particular class of drugs, both in terms of medical staff coordination and adapted infrastructures for efficient radioprotection.
Collapse
Affiliation(s)
- Emmanuel Deshayes
- INSERM U1194, Montpellier Cancer Research Institute, University of Montpellier, 34298 Montpellier, France
- Department of Nuclear Medicine, Institute du Cancer de Montpellier (ICM), 34298 Montpellier, France
| | - Cyril Fersing
- Department of Nuclear Medicine, Institute du Cancer de Montpellier (ICM), 34298 Montpellier, France
- IBMM, University Montpellier, CNRS, ENSCM, 34293 Montpellier, France
| | - Constance Thibault
- Department of Medical Oncology, Hôpital Européen Georges Pompidou, Institut du Cancer Paris CARPEM, AP-HP Centre, 75015 Paris, France
| | - Mathieu Roumiguie
- Urology Department, Andrology and Renal Transplantation, CHU Rangueil, 31059 Toulouse, France
| | - Philippe Pourquier
- INSERM U1194, Montpellier Cancer Research Institute, University of Montpellier, 34298 Montpellier, France
| | - Nadine Houédé
- INSERM U1194, Montpellier Cancer Research Institute, University of Montpellier, 34298 Montpellier, France
- Medical Oncology Department, Institute de Cancérologie du Gard-CHU Caremeau, 30009 Nîmes, France
| |
Collapse
|
16
|
Busslinger SD, Tschan VJ, Richard OK, Talip Z, Schibli R, Müller C. [ 225Ac]Ac-SibuDAB for Targeted Alpha Therapy of Prostate Cancer: Preclinical Evaluation and Comparison with [ 225Ac]Ac-PSMA-617. Cancers (Basel) 2022; 14:5651. [PMID: 36428743 PMCID: PMC9688344 DOI: 10.3390/cancers14225651] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
In the present study, SibuDAB, an albumin-binding PSMA ligand, was investigated in combination with actinium-225 and the data were compared with those of [225Ac]Ac-PSMA-617. In vitro, [225Ac]Ac-SibuDAB and [225Ac]Ac-PSMA-617 showed similar tumor cell uptake and PSMA-binding affinities as their 177Lu-labeled counterparts. The in vitro binding to serum albumin in mouse and human blood plasma, respectively, was 2.8-fold and 1.4-fold increased for [225Ac]Ac-SibuDAB as compared to [177Lu]Lu-SibuDAB. In vivo, this characteristic was reflected by the longer retention of [225Ac]Ac-SibuDAB in the blood than previously seen for [177Lu]Lu-SibuDAB. Similar to [225Ac]Ac-PSMA-617, [225Ac]Ac-SibuDAB was well tolerated at 30 kBq per mouse. Differences in blood cell counts were observed between treated mice and untreated controls, but no major variations were observed between values obtained for [225Ac]Ac-SibuDAB and [225Ac]Ac-PSMA-617. [225Ac]Ac-SibuDAB was considerably more effective to treat PSMA-positive tumor xenografts than [225Ac]Ac-PSMA-617. Only 5 kBq per mouse were sufficient to eradicate the tumors, whereas tumor regrowth was observed for mice treated with 5 kBq [225Ac]Ac-PSMA-617 and only one out of six mice survived until the end of the study. The enhanced therapeutic efficacy of [225Ac]Ac-SibuDAB as compared to that of [225Ac]Ac-PSMA-617 and reasonable safety data qualify this novel radioligand as a candidate for targeted α-therapy of prostate cancer.
Collapse
Affiliation(s)
- Sarah D. Busslinger
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland
| | - Viviane J. Tschan
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland
| | | | - Zeynep Talip
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, Vladimir-Prelog-Weg 1-5/10, ETH Zurich, 8093 Zurich, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, Vladimir-Prelog-Weg 1-5/10, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|