1
|
Cao W, Sergeeva O, Julian W, Kresak A, Lusinger D, Schneider J, Berridge MS, Sexton S, Wojtylak P, Li Q, Liu W, Chan ER, Saunthararajah Y, Lee Z. PET imaging of hepatocellular carcinoma with [ 124I]IV-14. EJNMMI Res 2025; 15:35. [PMID: 40192905 PMCID: PMC11977070 DOI: 10.1186/s13550-025-01227-5] [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: 01/06/2025] [Accepted: 03/16/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Currently, positron emission tomography (PET) plays no clear role in clinical imaging and management of hepatocellular carcinoma (HCC). New radiotracers for new target(s) are needed for PET imaging of HCC. Uridine-cytidine kinase 2 (UCK2) is a rate-limiting enzyme of the pyrimidine salvage synthesis pathway to phosphorylate uridine and cytidine. Studies have demonstrated that UCK2 is overexpressed in many types of solid cancers including HCC and is associated with the poor prognosis and proliferation of HCC. This study reported PET imaging using a UCK2-specific radiotracer with a clinically relevant anima model of spontaneously occurring HCC in the woodchucks. METHODS This study used 3'-(E)-(2-iodovinyl) uridine (IV-14), which is derived from a UCK2-selective antitumor agent 3'-(Ethynyl)uridine (EUrd), a cytotoxic ribonucleoside analogs of uridine. By radiolabeling IV-14 with Iodine-124 (124I), a UCK2-specific radiotracer [124I]IV-14 was obtained for PET imaging of UCK2. A naturally occurring woodchuck model of HCC following chronic viral hepatitis infection was used for PET imaging. Potassium iodide (KI) was tested in one of the three animals to block possible uptake of free 124I from de-iodination of [124I]IV-14. RESULTS We confirmed that UCK2 expression is higher in the woodchuck model of HCC than in the surrounding hepatic tissue, similar to human UCK2 that is highly expressed in human HCC. PET imaging with [124I]IV-14 showed a strong uptake in woodchuck HCC with low background uptake at one-hour post-injection. De-iodination did not seem to be an issue for PET imaging. CONCLUSION Our results demonstrate that UCK2 is a viable target for imaging HCC and has the potential for targeted endoradiotherapy of HCC.
Collapse
Affiliation(s)
- Wei Cao
- Radiology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Olga Sergeeva
- Radiology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - William Julian
- Radiology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Adam Kresak
- Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | | | | | | | - Sandra Sexton
- Medical Oncology, Roswell Park Cancer Institute, Buffalo, NY, 14263, USA
| | - Patrick Wojtylak
- Nuclear Medicine, Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Qiubai Li
- Nuclear Medicine, Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Wendy Liu
- Pathology, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Ernest Ricky Chan
- Cleveland Institute for Computational Biology, Cleveland, OH, 44106, USA
| | | | - Zhenghong Lee
- Radiology, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Nuclear Medicine, Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA.
- Radiology and Biomedical Engineering, Case Western Reserve University, 11100 Euclid Ave., Cleveland, OH, 44106, USA.
| |
Collapse
|
2
|
Fayn S, Roy S, Cabalteja CC, Lee W, Makala H, Baidoo K, Nambiar D, Sheehan‐Klenk J, Chung J, Buffington J, Ho M, Escorcia FE, Cheloha RW. Generation of Site-Specifically Labeled Affinity Reagents via Use of a Self-Labeling Single Domain Antibody. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417160. [PMID: 39965119 PMCID: PMC11984916 DOI: 10.1002/advs.202417160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/30/2025] [Indexed: 02/20/2025]
Abstract
Several chemical and enzymatic methods have been used to link antibodies to moieties that facilitate visualization of cognate targets. Emerging evidence suggests that the extent of labeling, dictated by the type of chemistry used, has a substantial impact on performance, especially in the context of antibodies used for the visualization of tumors in vivo. These effects are particularly pronounced in studies using small antibody fragments, such as single-domain antibodies, or nanobodies. Here, we leverage a new variety of conjugation chemistry, based on a nanobody that forms a crosslink with a specialized high-affinity epitope analogue, to label target-specific nanobody constructs with functionalities of choice, including fluorophores, chelators, and click chemistry handles. Using heterodimeric nanobody conjugates, comprised of an antigen recognition module and a self-labeling module, enables us to attach the desired functional group at a location distal to the site of antigen recognition. Constructs generated using this approach bound to antigens expressed on xenograft murine models of liver cancer and allowed for non-invasive diagnostic imaging. The modularity of our approach using a self-labeling nanobody offers a novel method for site-specific functionalization of biomolecules and can be extended to other applications for which covalent labeling is required.
Collapse
Affiliation(s)
- Stanley Fayn
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
- Oxford Institute for Radiation OncologyDepartment of OncologyUniversity of OxfordOxfordOX3 7DQUK
| | - Swarnali Roy
- Chemical Biology in Signaling SectionNational Institutes of Diabetes and Digestive and Kidney DiseasesNational Institutes of HealthBethesdaMD20892USA
| | - Chino C. Cabalteja
- Chemical Biology in Signaling SectionNational Institutes of Diabetes and Digestive and Kidney DiseasesNational Institutes of HealthBethesdaMD20892USA
| | - Woonghee Lee
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Hima Makala
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Kwamena Baidoo
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Divya Nambiar
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Julia Sheehan‐Klenk
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Joon‐Yong Chung
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Jesse Buffington
- Antibody Engineering ProgramCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Mitchell Ho
- Antibody Engineering ProgramCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
- Laboratory of Molecular BiologyCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Freddy E. Escorcia
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
- Radiation Oncology BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Ross W. Cheloha
- Chemical Biology in Signaling SectionNational Institutes of Diabetes and Digestive and Kidney DiseasesNational Institutes of HealthBethesdaMD20892USA
| |
Collapse
|
3
|
Tosato M, Favaretto C, Kleynhans J, Burgoyne AR, Gestin JF, van der Meulen NP, Jalilian A, Köster U, Asti M, Radchenko V. Alpha Atlas: Mapping global production of α-emitting radionuclides for targeted alpha therapy. Nucl Med Biol 2025; 142-143:108990. [PMID: 39809026 DOI: 10.1016/j.nucmedbio.2024.108990] [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: 11/09/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025]
Abstract
Targeted Alpha Therapy has shown great promise in cancer treatment, sparking significant interest over recent decades. However, its broad adoption has been impeded by the scarcity of alpha-emitters and the complexities related to their use. The availability of these radionuclides is often constrained by the intricate production processes and purification, as well as regulatory and logistical challenges. Moreover, the high cost and technical difficulties associated with handling and applying alpha-emitting radionuclides pose additional barriers to their clinical implementation. This Alpha Atlas provides an in-depth overview of the leading alpha-particle emitting radionuclide candidates for clinical use, focusing on their production processes and supply chains. By mapping the current facilities that produce and supply these radionuclides, this atlas aims to assist researchers, clinicians, and industries in initiating or scaling up the applications of alpha-emitters. The Alpha Atlas aspires to act as a strategic guide, facilitating collaboration and driving forward the integration of these potent therapeutic agents into cancer treatment practices.
Collapse
Affiliation(s)
- Marianna Tosato
- Radiopharmaceutical Chemistry Laboratory (RACHEL), Nuclear Medicine Unit, AUSL-IRCCS Reggio Emilia, 42123 Reggio Emilia, Italy.
| | - Chiara Favaretto
- Radiopharmacy and Cyclotron Department, IRCCS Sacro Cuore Don Calabria, Negrar 37024, Verona, Italy
| | - Janke Kleynhans
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Andrew R Burgoyne
- Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, TN 37830, United States
| | - Jean-François Gestin
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44000 Nantes, France
| | - Nicholas P van der Meulen
- PSI Center for Life Sciences, 5232 Villigen-PSI, Switzerland; PSI Center for Nuclear Engineering and Sciences, 5232 Villigen-PSI, Switzerland
| | - Amirreza Jalilian
- Department of Nuclear Safety and Security, International Atomic Energy Agency, 1220 Vienna, Austria
| | - Ulli Köster
- Institut Laue-Langevin, 38042 Grenoble, France
| | - Mattia Asti
- Radiopharmaceutical Chemistry Laboratory (RACHEL), Nuclear Medicine Unit, AUSL-IRCCS Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, BC V6T 2A3 Vancouver, British Columbia, Canada; Department of Chemistry, University of British Columbia, V6T 1Z1 Vancouver, British Columbia, Canada
| |
Collapse
|
4
|
Tang L, Yang X, He L, Zhu C, Chen Q. Preclinical advance in nanoliposome-mediated photothermal therapy in liver cancer. Lipids Health Dis 2025; 24:31. [PMID: 39891269 PMCID: PMC11783920 DOI: 10.1186/s12944-024-02429-x] [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: 10/23/2024] [Accepted: 12/31/2024] [Indexed: 02/03/2025] Open
Abstract
Liver cancer is a highly lethal malignant tumor with a high incidence worldwide. Therefore, its treatment has long been a focus of medical research. Although traditional treatment methods such as surgery, radiotherapy, and chemotherapy have increased the survival rate of patients, their efficacy remains unsatisfactory owing to the nonspecific distribution of drugs, high toxicity, and drug resistance of tumor tissues. In recent years, the application of nanotechnology in the medical field has opened a new avenue for the treatment of liver cancer. Among these treatment methods, photothermal therapy (PTT) based on nanoliposomes has attracted wide attention owing to its unique targeting and high efficiency. This article reviews the latest preclinical research progress of nanoliposome-based PTT for liver cancer and its metastasis, discusses the preclinical challenges in this field, and proposes directions for improvement, with the aim of improving the effectiveness of liver cancer treatment.
Collapse
Affiliation(s)
- Lixuan Tang
- School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Xiao Yang
- The department of oncology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Liwen He
- School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Chaogeng Zhu
- The department of hepatobiliary pancreatic hernia surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Qingshan Chen
- The department of hepatobiliary pancreatic hernia surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
5
|
Woods JJ, Rigby A, Wacker JN, Arino T, Alvarenga Vasquez JV, Cosby A, Martin KE, Abergel RJ. Synthesis and Evaluation of a Bifunctional Chelator for Thorium-227 Targeted Radiotherapy. J Med Chem 2025; 68:1682-1692. [PMID: 39752149 DOI: 10.1021/acs.jmedchem.4c02423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Thorium-227 (227Th) is an α-emitting radionuclide currently under investigation for targeted alpha therapy. Available chelators used for this isotope suffer from challenging multistep syntheses. Here, we present the synthesis and preclinical evaluation of a novel bifunctional chelator, p-SCN-Bn-DOTHOPO, which contains an isothiocyanate group that is suitable for conjugation to biological molecules. This bifunctional chelator was prepared with a 26% overall yield in four steps and conjugated to the human epidermal growth factor receptor 2 targeting antibody, trastuzumab. The resulting immunoconjugate was labeled with [227Th]ThIV (pH 5.5, room temperature, 60 min) with ≥95% radiochemical yield and purity. The conjugate was also labeled with zirconium-89 (89Zr), which can be used for positron emission tomography imaging. The radiometal complexes were subsequently investigated for their biological stability. The results described here provide insight into ligand design strategies and optimization of chelators for the development of the next generation of 89Zr and 227Th radiopharmaceuticals.
Collapse
Affiliation(s)
- Joshua J Woods
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Alex Rigby
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Jennifer N Wacker
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Trevor Arino
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
- Department of Nuclear Engineering, University of California Berkeley, Berkeley, California 94720, United States
| | | | - Alexia Cosby
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Kirsten E Martin
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Rebecca J Abergel
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
- Department of Nuclear Engineering, University of California Berkeley, Berkeley, California 94720, United States
- Department of Chemistry, University of California Berkeley, Berkeley, California 94720, United States
| |
Collapse
|
6
|
Yang L, Pham K, Xi Y, Wu Q, Liu D, Robertson KD, Liu C. Exploring Glypican-3 targeted CAR-NK treatment and potential therapy resistance in hepatocellular carcinoma. PLoS One 2025; 20:e0317401. [PMID: 39841705 PMCID: PMC11753693 DOI: 10.1371/journal.pone.0317401] [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: 06/11/2024] [Accepted: 12/27/2024] [Indexed: 01/24/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent form of primary liver cancer and the second leading cause of cancer-related mortality globally. Despite advancements in current HCC treatment, it remains a malignancy with poor prognosis. Therefore, developing novel treatment options for patients with HCC is urgently needed. Chimeric antigen receptor (CAR)-modified natural killer (NK) cells have demonstrated potent anti-tumor effects, making them as a promising immunotherapy strategy for cancer treatment. Glypican-3 (GPC3), a cell surface oncofetal glycoprotein, is highly expressed in most HCC tissues, but not in normal tissues, and functions as a key driver of carcinogenesis. Given its high expression level on the cell surface, GPC3 is considered as an attractive immunotherapy target for HCC. In this study, two GPC3-specific CAR-NK cells, NK92MI/HN3 and NK92MI/HS20, were established using NK92MI cells, a modified IL-2-independent NK cell line. These cell lines were engineered with third generation GPC3-specific CARs, and their activities were subsequently evaluated in the treatment of HCC. We found that NK92MI/HN3 cells, rather than NK92MI/HS20 cells, exhibited a significant cytotoxicity effect against GPC3+ HepG2 cells in vitro and efficiently suppressed tumor growth in a xenograft model using NSG mice. In addition, irradiated NK92MI/HN3 cells displayed similar anti-tumor efficacy to unirradiated NK92MI/HN3 cells. Furthermore, we observed that NK92MI/HN3 cells showed higher killing activity against the GPC3 isoform 2 overexpression cell line (Sk-Hep1-v2) than those with GPC3 isoform 1 overexpression cell line (Sk-Hep1-v1). This suggest that the presence of different GPC3 isoforms in HCC may impact the cytotoxicity activity of NK92MI/HN3 cells and potentially influence therapeutic outcomes. These findings highlight the effective anti-HCC effects of NK92MI/HN3 cells and reveal the role of GPC3 isoforms in influencing therapy outcomes, suggesting that isoform analysis should be considered to optimize CAR-NK therapies to improve patient outcomes.
Collapse
MESH Headings
- Glypicans/immunology
- Glypicans/metabolism
- Glypicans/antagonists & inhibitors
- Carcinoma, Hepatocellular/therapy
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Liver Neoplasms/therapy
- Liver Neoplasms/immunology
- Liver Neoplasms/pathology
- Humans
- Animals
- Killer Cells, Natural/immunology
- Killer Cells, Natural/transplantation
- Killer Cells, Natural/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Mice
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Immunotherapy, Adoptive/methods
Collapse
Affiliation(s)
- Lei Yang
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut, United States of America
| | - Kien Pham
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut, United States of America
| | - Yibo Xi
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut, United States of America
| | - Qunfeng Wu
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Dongfang Liu
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Keith D. Robertson
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Chen Liu
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut, United States of America
| |
Collapse
|
7
|
Li Z, Mo C, Li C, Wang Q, Huang S, Huang Y, Liang Y. Gallium-68 Labeled Positron Emission Computed Tomography Tracer Targeting Glypican-3 with High Contrast for Hepatocellular Carcinoma Imaging. ACS Pharmacol Transl Sci 2024; 7:4021-4031. [PMID: 39698271 PMCID: PMC11651169 DOI: 10.1021/acsptsci.4c00504] [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: 08/19/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024]
Abstract
Hepatocellular carcinoma (HCC) represents the predominant form of primary liver cancer, yet early, precise, and noninvasive detection continues to pose a considerable clinical challenge. Glypican-3 (GPC3), a membrane-bound proteoglycan, is markedly overexpressed in most HCC cases, while exhibiting low expression in normal and hepatitis-affected liver tissues. Given its crucial role in malignant transformation and tumor progression, GPC3 emerges as a compelling target for imaging. In this study, we developed and evaluated 2 68Ga-labeled GPC3-targeted positron emission tomography (PET) probes, each incorporating either polyethylene glycol (PEG) or 4-(p-methylphenyl)butanoic acid (an albumin-binding moiety). Comparative analyses revealed that 68Ga-ALB-GBP, which includes the albumin-binding moiety, exhibited superior in vivo stability, enhanced tumor uptake, and an improved tumor-to-liver ratio relative to 68Ga-PEG2-GBP in subcutaneous HCC mouse models. Micro-PET/computed tomography imaging of orthotopic liver cancer with 68Ga-ALB-GBP demonstrated a tumor-to-liver ratio of 2.29 ± 0.13 and a tumor-to-muscle ratio of 13.03 ± 1.63 at 3 h postinjection, outperforming the performance of the clinically used 18F-fluorodeoxyglucose PET imaging. These findings suggest that 68Ga-ALB-GBP is a promising diagnostic tool for HCC and a strong candidate for clinical translation with potential utility in both diagnostic and therapeutic settings. Moreover, the incorporation of an albumin-binding moiety into PET tracers significantly extends blood circulation time, thereby enhancing bioavailability and facilitating high-contrast PET imaging.
Collapse
Affiliation(s)
- Zhongjing Li
- Department
of Nuclear Medicine, National Cancer Center/National Clinical Research
Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union
Medical College, Shenzhen 518116, China
| | - Chunwei Mo
- Department
of Nuclear Medicine, Nanfang Hospital, GDMPA Key Laboratory for Quality
Control and Evaluation of Radiopharmaceuticals, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Chengzhe Li
- Department
of Nuclear Medicine, National Cancer Center/National Clinical Research
Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union
Medical College, Shenzhen 518116, China
| | - Qiong Wang
- Department
of Nuclear Medicine, National Cancer Center/National Clinical Research
Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union
Medical College, Shenzhen 518116, China
| | - Size Huang
- Department
of Nuclear Medicine, National Cancer Center/National Clinical Research
Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union
Medical College, Shenzhen 518116, China
| | - Yong Huang
- Department
of Nuclear Medicine, National Cancer Center/National Clinical Research
Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union
Medical College, Shenzhen 518116, China
| | - Ying Liang
- Department
of Nuclear Medicine, National Cancer Center/National Clinical Research
Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union
Medical College, Shenzhen 518116, China
| |
Collapse
|
8
|
Zuo D, Wang H, Yu B, Li Q, Gan L, Chen W. Astatine-211 and actinium-225: two promising nuclides in targeted alpha therapy. Acta Biochim Biophys Sin (Shanghai) 2024; 57:327-343. [PMID: 39587859 PMCID: PMC11986457 DOI: 10.3724/abbs.2024206] [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: 07/11/2024] [Accepted: 10/22/2024] [Indexed: 11/27/2024] Open
Abstract
Nuclear medicine therapy offers a promising approach for tumor treatment, as the energy emitted during radionuclide decay causes irreparable damage to tumor cells. Notably, α-decay exhibits an even more significant destructive potential. By conjugating α-nuclides with antibodies or small-molecule inhibitors, targeted alpha therapy (TAT) can enhance tumor destruction while minimizing toxic side effects, making TAT an increasingly attractive antineoplastic strategy. Astatine-211 ( 211At) and actinium-225 ( 225Ac) have emerged as highly effective agents in TAT due to their exceptional physicochemical properties and biological effects. In this review, we highlight the applications of 211At-/ 225Ac-radiopharmaceuticals, particularly in specific tumor targets, such as prostate-specific membrane antigen (PSMA) in prostate cancers, cluster of differentiation (CD) in hematological malignancies, human epidermal growth factor receptor-2 (HER2) in ovarian cancers, and somatostatin receptor (SSTR) in neuroendocrine tumors. We synthesize the progress from preclinical and clinical trials to provide insights into the promising potential of 211At-/ 225Ac-radiopharmaceuticals for future treatments.
Collapse
Affiliation(s)
- Dashan Zuo
- Institute of Modern PhysicsChinese Academy of SciencesLanzhou730000China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhou730000China
- University of Chinese Academy of SciencesBeijing100049China
| | - Hui Wang
- Institute of Modern PhysicsChinese Academy of SciencesLanzhou730000China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhou730000China
| | - Boyi Yu
- Institute of Modern PhysicsChinese Academy of SciencesLanzhou730000China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhou730000China
| | - Qiang Li
- Institute of Modern PhysicsChinese Academy of SciencesLanzhou730000China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhou730000China
- University of Chinese Academy of SciencesBeijing100049China
- Lanhai Nuclear Medical Research CenterPutian351153China
| | - Lu Gan
- Institute of Modern PhysicsChinese Academy of SciencesLanzhou730000China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhou730000China
- University of Chinese Academy of SciencesBeijing100049China
| | - Weiqiang Chen
- Institute of Modern PhysicsChinese Academy of SciencesLanzhou730000China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhou730000China
- University of Chinese Academy of SciencesBeijing100049China
- Lanhai Nuclear Medical Research CenterPutian351153China
| |
Collapse
|
9
|
Gharibkandi NA, Wawrowicz K, Walczak R, Majkowska-Pilip A, Wierzbicki M, Bilewicz A. 109Pd/ 109mAg in-vivo generator in the form of nanoparticles for combined β - - Auger electron therapy of hepatocellular carcinoma. EJNMMI Radiopharm Chem 2024; 9:59. [PMID: 39136900 PMCID: PMC11322470 DOI: 10.1186/s41181-024-00293-9] [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: 06/13/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Convenient therapeutic protocols for hepatocellular carcinoma (HCC) are often ineffective due to late diagnosis and high tumor heterogeneity, leading to poor long-term outcomes. However, recently performed studies suggest that using nanostructures in liver cancer treatment may improve therapeutic effects. Inorganic nanoparticles represent a unique material that tend to accumulate in the liver when introduced in-vivo. Typically, this is a major drawback that prevents the therapeutic use of nanoparticles in medicine. However, in HCC tumours, this may be advantageous because nanoparticles may accumulate in the target organ, where the leaky vasculature of HCC causes their accumulation in tumour cells via the EPR effect. On the other hand, recent studies have shown that combining low- and high-LET radiation emitted from the same radionuclide, such as 161Tb, can increase the effectiveness of radionuclide therapy. Therefore, to improve the efficacy of radionuclide therapy for hepatocellular carcinoma, we suggest utilizing radioactive palladium nanoparticles in the form of 109Pd/109mAg in-vivo generator that simultaneously emits β- particles and Auger electrons. RESULTS Palladium nanoparticles with a size of 5 nm were synthesized using 109Pd produced through neutron irradiation of natural palladium or enriched 108Pd. Unlike the 109Pd-cyclam complex, where the daughter radionuclide diffuses away from the molecules, 109mAg remains within the nanoparticles after the decay of 109Pd. In vitro cell studies using radioactive 109Pd nanoparticles revealed that the nanoparticles accumulated inside cells, reaching around 50% total uptake. The 109Pd-PEG nanoparticles exhibited high cytotoxicity, even at low levels of radioactivity (6.25 MBq/mL), resulting in almost complete cell death at 25 MBq/mL. This cytotoxic effect was significantly greater than that of PdNPs labeled with β- (131I) and Auger electron emitters (125I). The metabolic viability of HCC cells was found to be correlated with cell DNA DSBs. Also, successful radioconjugate anticancer activity was observed in three-dimensional tumor spheroids, resulting in a significant treatment response. CONCLUSION The results indicate that nanoparticles labeled with 109Pd can be effectively used for combined β- - Auger electron-targeted radionuclide therapy of HCC. Due to the decay of both components (β- and Auger electrons), the 109Pd/109mAg in-vivo generator presents a unique potential in this field.
Collapse
Affiliation(s)
- Nasrin Abbasi Gharibkandi
- Centre of Radiochemistry and Nuclear Chemistry, Institute of Nuclear Chemistry and Technology, Dorodna 16 St, Warsaw, 03-195, Poland
| | - Kamil Wawrowicz
- Department of Medical Physics, M. Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Rafał Walczak
- Centre of Radiochemistry and Nuclear Chemistry, Institute of Nuclear Chemistry and Technology, Dorodna 16 St, Warsaw, 03-195, Poland
| | - Agnieszka Majkowska-Pilip
- Centre of Radiochemistry and Nuclear Chemistry, Institute of Nuclear Chemistry and Technology, Dorodna 16 St, Warsaw, 03-195, Poland.
- Department of Nuclear Medicine, National Medical Institute of the Ministry of the Interior and Administration, Wołoska 137 St, Warsaw, 02-507, Poland.
| | - Mateusz Wierzbicki
- Institute of Biology, Warsaw University of Life Sciences, Ciszewskiego 8 St, Warsaw, 02-786, Poland
| | - Aleksander Bilewicz
- Centre of Radiochemistry and Nuclear Chemistry, Institute of Nuclear Chemistry and Technology, Dorodna 16 St, Warsaw, 03-195, Poland.
| |
Collapse
|
10
|
Yang Y, Li P, Feng H, Zeng R, Li S, Zhang Q. Macrocycle-Based Supramolecular Drug Delivery Systems: A Concise Review. Molecules 2024; 29:3828. [PMID: 39202907 PMCID: PMC11357536 DOI: 10.3390/molecules29163828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/26/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
Efficient delivery of therapeutic agents to the lesion site or specific cells is an important way to achieve "toxicity reduction and efficacy enhancement". Macrocycles have always provided many novel ideas for drug or gene loading and delivery processes. Specifically, macrocycles represented by crown ethers, cyclodextrins, cucurbit[n]urils, calix[n]arenes, and pillar[n]arenes have unique properties, which are different cavity structures, good biocompatibility, and good stability. Benefited from these diverse properties, a variety of supramolecular drug delivery systems can be designed and constructed to effectively improve the physical and chemical properties of guest molecules as needed. This review provides an outlook on the current application status and main limitations of macrocycles in supramolecular drug delivery systems.
Collapse
Affiliation(s)
- Yanrui Yang
- College of Pharmacy, Key Laboratory of Research and Application of Ethnic Medicine Processing and Preparation on the Qinghai Tibet Plateau, Southwest Minzu University, Chengdu 610041, China
| | - Pengcheng Li
- College of Pharmacy, Key Laboratory of Research and Application of Ethnic Medicine Processing and Preparation on the Qinghai Tibet Plateau, Southwest Minzu University, Chengdu 610041, China
| | - Haibo Feng
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China
| | - Rui Zeng
- College of Pharmacy, Key Laboratory of Research and Application of Ethnic Medicine Processing and Preparation on the Qinghai Tibet Plateau, Southwest Minzu University, Chengdu 610041, China
| | - Shanshan Li
- College of Pharmacy, Key Laboratory of Research and Application of Ethnic Medicine Processing and Preparation on the Qinghai Tibet Plateau, Southwest Minzu University, Chengdu 610041, China
| | - Qixiong Zhang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Department of Pharmacy, Sichuan Provincial People’s Hospital Chuandong Hospital & Dazhou First People’s Hospital, Dazhou 635000, China
| |
Collapse
|
11
|
Chung JY, Lee W, Lee OW, Ylaya K, Nambiar D, Sheehan-Klenk J, Fayn S, Hewitt SM, Choyke PL, Escorcia FE. Glypican-3 deficiency in liver cancer upregulates MAPK/ERK pathway but decreases cell proliferation. Am J Cancer Res 2024; 14:3348-3371. [PMID: 39113871 PMCID: PMC11301284 DOI: 10.62347/ttny4279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/20/2024] [Indexed: 08/10/2024] Open
Abstract
Glypican-3 (GPC3) is overexpressed in hepatocellular carcinomas and hepatoblastomas and represents an important therapeutic target but the biologic importance of GPC3 in liver cancer is unclear. To date, there are limited data characterizing the biological implications of GPC3 knockout (KO) in liver cancers that intrinsically express this target. Here, we report on the development and characterization of GPC3-KO liver cancer cell lines and compare to them to parental lines. GPC3-KO variants were established in HepG2 and Hep3B liver cancer cell lines using a lentivirus-mediated CRISPR/Cas9 system. We assessed the effects of GPC3 deficiency on oncogenic properties in vitro and in murine xenograft models. Downstream cellular signaling pathway changes induced by GPC3 deficiency were examined by RNAseq and western blot. To confirm the usefulness of the models for GPC3-targeted drug development, we evaluated the target engagement of a GPC3-selective antibody, GC33, conjugated to the positron-emitting zirconium-89 (89Zr) in subcutaneous murine xenografts of wild type (WT) and KO liver cancer cell lines. Deletion of GPC3 significantly reduced liver cancer cell proliferation, migration, and invasion compared to the parental cell lines. Additionally, the tumor growth of GPC3-KO liver cancer xenografts was significantly slower compared with control xenografts. RNA sequencing analysis also showed GPC3-KO resulted in a reduction in the expression of genes associated with cell cycle regulation, invasion, and migration. Specifically, we observed the downregulation of components in the AKT/NFκB/WNT signaling pathways and of molecules related to cell cycle regulation with GPC3-KO. In contrast, pMAPK/ERK1/2 was upregulated, suggesting an adaptive compensatory response. KO lines demonstrated increased sensitivity to ERK (GDC09994), while AKT (MK2206) inhibition was more effective in WT lines. Using antibody-based positron emission tomography (immunoPET) imaging, we confirmed that 89Zr-GC33 accumulated exclusively in GPC3-expression xenografts but not in GPC3-KO xenografts with high tumor uptake and tumor-to-liver signal ratio. We show that GPC3-KO liver cancer cell lines exhibit decreased tumorigenicity and altered signaling pathways, including upregulated pMAPK/ERK1/2, compared to parental lines. Furthermore, we successfully distinguished between GPC3+ and GPC3- tumors using the GPC3-targeted immunoPET imaging agent, demonstrating the potential utility of these cell lines in facilitating GPC3-selective drug development.
Collapse
Affiliation(s)
- Joon-Yong Chung
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| | - Woonghee Lee
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| | - Olivia W Lee
- Laboratory of Genetic Susceptibility, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| | - Kris Ylaya
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| | - Divya Nambiar
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| | - Julia Sheehan-Klenk
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| | - Stanley Fayn
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
- Oxford Institute for Radiation Oncology, Department of Oncology, University of OxfordOxford OX3 7DQ, UK
| | - Stephen M Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| | - Peter L Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| | - Freddy E Escorcia
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| |
Collapse
|
12
|
Lin F, Clift R, Ehara T, Yanagida H, Horton S, Noncovich A, Guest M, Kim D, Salvador K, Richardson S, Miller T, Han G, Bhat A, Song K, Li G. Peptide Binder to Glypican-3 as a Theranostic Agent for Hepatocellular Carcinoma. J Nucl Med 2024; 65:586-592. [PMID: 38423788 DOI: 10.2967/jnumed.123.266766] [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: 10/16/2023] [Revised: 01/23/2024] [Indexed: 03/02/2024] Open
Abstract
Glypican-3 (GPC3) is a membrane-associated glycoprotein that is significantly upregulated in hepatocellular carcinomas (HCC) with minimal to no expression in normal tissues. The differential expression of GPC3 between tumor and normal tissues provides an opportunity for targeted radiopharmaceutical therapy to treat HCC, a leading cause of cancer-related deaths worldwide. Methods: DOTA-RYZ-GPC3 (RAYZ-8009) comprises a novel macrocyclic peptide binder to GPC3, a linker, and a chelator that can be complexed with different radioisotopes. The binding affinity was determined by surface plasma resonance and radioligand binding assays. Target-mediated cellular internalization was radiometrically measured at multiple time points. In vivo biodistribution, monotherapy, and combination treatments with 177Lu or 225Ac were performed on HCC xenografts. Results: RAYZ-8009 showed high binding affinity to GPC3 protein of human, mouse, canine, and cynomolgus monkey origins and no binding to other glypican family members. Potent cellular binding was confirmed in GPC3-positive HepG2 cells and was not affected by isotope switching. RAYZ-8009 achieved efficient internalization on binding to HepG2 cells. Biodistribution study of 177Lu-RAYZ-8009 showed sustained tumor uptake and fast renal clearance, with minimal or no uptake in other normal tissues. Tumor-specific uptake was also demonstrated in orthotopic HCC tumors, with no uptake in surrounding liver tissue. Therapeutically, significant and durable tumor regression and survival benefit were achieved with 177Lu- and 225Ac-labeled RAYZ-8009, as single agents and in combination with lenvatinib, in GPC3-positive HCC xenografts. Conclusion: Preclinical in vitro and in vivo data demonstrate the potential of RAYZ-8009 as a theranostic agent for the treatment of patients with GPC3-positive HCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Matt Guest
- RayzeBio, Inc., San Diego, California; and
| | - Daniel Kim
- RayzeBio, Inc., San Diego, California; and
| | | | | | | | | | | | | | - Gary Li
- RayzeBio, Inc., San Diego, California; and
| |
Collapse
|
13
|
Hooijman EL, Radchenko V, Ling SW, Konijnenberg M, Brabander T, Koolen SLW, de Blois E. Implementing Ac-225 labelled radiopharmaceuticals: practical considerations and (pre-)clinical perspectives. EJNMMI Radiopharm Chem 2024; 9:9. [PMID: 38319526 PMCID: PMC10847084 DOI: 10.1186/s41181-024-00239-1] [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: 07/20/2023] [Accepted: 01/25/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND In the past years, there has been a notable increase in interest regarding targeted alpha therapy using Ac-225, driven by the observed promising clinical anti-tumor effects. As the production and technology has advanced, the availability of Ac-225 is expected to increase in the near future, making the treatment available to patients worldwide. MAIN BODY Ac-225 can be labelled to different biological vectors, whereby the success of developing a radiopharmaceutical depends heavily on the labelling conditions, purity of the radionuclide source, chelator, and type of quenchers used to avoid radiolysis. Multiple (methodological) challenges need to be overcome when working with Ac-225; as alpha-emission detection is time consuming and highly geometry dependent, a gamma co-emission is used, but has to be in equilibrium with the mother-nuclide. Because of the high impact of alpha emitters in vivo it is highly recommended to cross-calibrate the Ac-225 measurements for used quality control (QC) techniques (radio-TLC, HPLC, HP-Ge detector, and gamma counter). More strict health physics regulations apply, as Ac-225 has a high toxicity, thereby limiting practical handling and quantities used for QC analysis. CONCLUSION This overview focuses specifically on the practical and methodological challenges when working with Ac-225 labelled radiopharmaceuticals, and underlines the required infrastructure and (detection) methods for the (pre-)clinical application.
Collapse
Affiliation(s)
- Eline L Hooijman
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 CN, Rotterdam, The Netherlands
- Department of Hospital Pharmacy, Erasmus MC, 3015 CN, Rotterdam, The Netherlands
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, Vancouver, BC, V6T 2A3, Canada
- Chemistry Department, University of British Columbia, Vancouver, BC, V6T 1Z1, Canada
| | - Sui Wai Ling
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 CN, Rotterdam, The Netherlands
| | - Mark Konijnenberg
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 CN, Rotterdam, The Netherlands
| | - Tessa Brabander
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 CN, Rotterdam, The Netherlands
| | - Stijn L W Koolen
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 CN, Rotterdam, The Netherlands
- Department of Hospital Pharmacy, Erasmus MC, 3015 CN, Rotterdam, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 CN, Rotterdam, The Netherlands
| | - Erik de Blois
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 CN, Rotterdam, The Netherlands.
| |
Collapse
|
14
|
Hassan M, Bokhari TH, Lodhi NA, Khosa MK, Usman M. A review of recent advancements in Actinium-225 labeled compounds and biomolecules for therapeutic purposes. Chem Biol Drug Des 2023; 102:1276-1292. [PMID: 37715360 DOI: 10.1111/cbdd.14311] [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: 03/16/2022] [Revised: 06/03/2023] [Accepted: 07/17/2023] [Indexed: 09/17/2023]
Abstract
In nuclear medicine, cancers that cannot be cured or can only be treated partially by traditional techniques like surgery or chemotherapy are killed by ionizing radiation as a form of therapeutic treatment. Actinium-225 is an alpha-emitting radionuclide that is highly encouraging as a therapeutic approach and more promising for targeted alpha therapy (TAT). Actinium-225 is the best candidate for tumor cells treatment and has physical characteristics such as high (LET) linear energy transfer (150 keV per μm), half-life (t1/2 = 9.92d), and short ranges (400-100 μm) which prevent the damage of normal healthy tissues. The introduction of various new radiopharmaceuticals and radioisotopes has significantly assisted the advancement of nuclear medicine. Ac-225 radiopharmaceuticals continuously demonstrate their potential as targeted alpha therapeutics. 225 Ac-labeled radiopharmaceuticals have confirmed their importance in medical and clinical areas by introducing [225 Ac]Ac-PSMA-617, [225 Ac]Ac-DOTATOC, [225 Ac]Ac-DOTA-substance-P, reported significantly improved response in patients with prostate cancer, neuroendocrine, and glioma, respectively. The development of these radiopharmaceuticals required a suitable buffer, incubation time, optimal pH, and reaction temperature. There is a growing need to standardize quality control (QC) testing techniques such as radiochemical purity (RCP). This review aims to summarize the development of the Ac-225 labeled compounds and biomolecules. The current state of their reported resulting clinical applications is also summarized as well.
Collapse
Affiliation(s)
- Maria Hassan
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| | | | - Nadeem Ahmed Lodhi
- Isotope Production Division, Pakistan institute of Nuclear Science & Technology (PINSTECH), Islamabad, Pakistan
| | | | - Muhammad Usman
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| |
Collapse
|
15
|
Njenga LW, Mbugua SN, Odhiambo RA, Onani MO. Addressing the gaps in homeostatic mechanisms of copper and copper dithiocarbamate complexes in cancer therapy: a shift from classical platinum-drug mechanisms. Dalton Trans 2023; 52:5823-5847. [PMID: 37021641 DOI: 10.1039/d3dt00366c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
The platinum drug, cisplatin, is considered as among the most successful medications in cancer treatment. However, due to its inherent toxicity and resistance limitations, research into other metal-based non-platinum anticancer medications with diverse mechanisms of action remains an active field. In this regard, copper complexes feature among non-platinum compounds which have shown promising potential as effective anticancer drugs. Moreover, the interesting discovery that cancer cells can alter their copper homeostatic processes to develop resistance to platinum-based treatments leads to suggestions that some copper compounds can indeed re-sensitize cancer cells to these drugs. In this work, we review copper and copper complexes bearing dithiocarbamate ligands which have shown promising results as anticancer agents. Dithiocarbamate ligands act as effective ionophores to convey the complexes of interest into cells thereby influencing the metal homeostatic balance and inducing apoptosis through various mechanisms. We focus on copper homeostasis in mammalian cells and on our current understanding of copper dysregulation in cancer and recent therapeutic breakthroughs using copper coordination complexes as anticancer drugs. We also discuss the molecular foundation of the mechanisms underlying their anticancer action. The opportunities that exist in research for these compounds and their potential as anticancer agents, especially when coupled with ligands such as dithiocarbamates, are also reviewed.
Collapse
Affiliation(s)
- Lydia W Njenga
- Department of Chemistry, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya.
| | - Simon N Mbugua
- Department of Chemistry, Kisii University, P.O. Box 408-40200, Kisii, Kenya
| | - Ruth A Odhiambo
- Department of Chemistry, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya.
| | - Martin O Onani
- Department of Chemical Sciences, University of the Western Cape, Private Bag X17, Belville, 7535, South Africa
| |
Collapse
|
16
|
Labadie KP, Lehnert AL, Kenoyer AL, Hamlin DK, Ludwig AD, Utria AF, Daniel SK, Mihailovic TN, Prossnitz A, Orozco JJ, Li Y, Wilbur DS, Miyaoka RS, Park JO. Glypican-3 targeted positron emission tomography detects sub-centimeter tumors in a xenograft model of hepatocellular carcinoma. EJNMMI Res 2023; 13:35. [PMID: 37103671 PMCID: PMC10140215 DOI: 10.1186/s13550-023-00980-9] [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: 02/15/2023] [Accepted: 03/29/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND Early intrahepatic recurrence is common after surgical resection of hepatocellular carcinoma (HCC) and leads to increased morbidity and mortality. Insensitive and nonspecific diagnostic imaging contributes to EIR and results in missed treatment opportunities. In addition, novel modalities are needed to identify targets amenable for targeted molecular therapy. In this study, we evaluated a zirconium-89 radiolabeled glypican-3 (GPC3) targeting antibody conjugate (89Zr-αGPC3) for use in positron emission tomography (PET) for detection of small, GPC3+ HCC in an orthotopic murine model. Athymic nu/J mice received hepG2, a GPC3+ human HCC cell line, into the hepatic subcapsular space. Tumor-bearing mice were imaged by PET/computerized tomography (CT) 4 days after tail vein injection of 89Zr-αGPC3. Livers were then excised for the tumors to be identified, measured, bisected, and then serially sectioned at 500 μm increments. Sensitivity and specificity of PET/CT for 89Zr-αGPC3-avid tumors were assessed using tumor confirmation on histologic sections as the gold standard. RESULTS In tumor-bearing mice, 89Zr-αGPC3 avidly accumulated in the tumor within four hours of injection with ongoing accumulation over time. There was minimal off-target deposition and rapid bloodstream clearance. Thirty-eight of 43 animals had an identifiable tumor on histologic analysis. 89Zr-αGPC3 immuno-PET detected all 38 histologically confirmed tumors with a sensitivity of 100%, with the smallest tumor detected measuring 330 μm in diameter. Tumor-to-liver ratios of 89Zr-αGPC3 uptake were high, creating excellent spatial resolution for ease of tumor detection on PET/CT. Two of five tumors that were observed on PET/CT were not identified on histologic analysis, yielding a specificity of 60%. CONCLUSIONS 89Zr-αGPC3 avidly accumulated in GPC3+ tumors with minimal off-target sequestration. 89Zr-αGPC3 immuno-PET yielded a sensitivity of 100% and detected sub-millimeter tumors. This technology may improve diagnostic sensitivity of small HCC and select GPC3+ tumors for targeted therapy. Human trials are warranted to assess its impact.
Collapse
Affiliation(s)
- Kevin P Labadie
- Department of Surgery, University of Washington School of Medicine, 1959 NE Pacific Street, Health Sciences Bldg. Room BB-442, Box 356410, Seattle, WA, 98195-6410, USA
| | - Adrienne L Lehnert
- Department of Radiology, University of Washington School of Medicine, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Aimee L Kenoyer
- Clinical Research Division, Fred Hutch Cancer Research Center, 100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Donald K Hamlin
- Department of Radiation Oncology, University of Washington School of Medicine, 616 NE Northlake Pl., Seattle, WA, 98105, USA
| | - Andrew D Ludwig
- Department of Surgery, University of Washington School of Medicine, 1959 NE Pacific Street, Health Sciences Bldg. Room BB-442, Box 356410, Seattle, WA, 98195-6410, USA
| | - Alan F Utria
- Department of Surgery, University of Washington School of Medicine, 1959 NE Pacific Street, Health Sciences Bldg. Room BB-442, Box 356410, Seattle, WA, 98195-6410, USA
| | - Sara K Daniel
- Department of Surgery, University of Washington School of Medicine, 1959 NE Pacific Street, Health Sciences Bldg. Room BB-442, Box 356410, Seattle, WA, 98195-6410, USA
| | - Tara N Mihailovic
- Department of Surgery, University of Washington School of Medicine, 1959 NE Pacific Street, Health Sciences Bldg. Room BB-442, Box 356410, Seattle, WA, 98195-6410, USA
| | - Alexander Prossnitz
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, WA, 98195, USA
| | - Johnnie J Orozco
- Clinical Research Division, Fred Hutch Cancer Research Center, 100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Yawen Li
- Department of Radiation Oncology, University of Washington School of Medicine, 616 NE Northlake Pl., Seattle, WA, 98105, USA
| | - D Scott Wilbur
- Department of Radiation Oncology, University of Washington School of Medicine, 616 NE Northlake Pl., Seattle, WA, 98105, USA
| | - Robert S Miyaoka
- Department of Radiology, University of Washington School of Medicine, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - James O Park
- Department of Surgery, University of Washington School of Medicine, 1959 NE Pacific Street, Health Sciences Bldg. Room BB-442, Box 356410, Seattle, WA, 98195-6410, USA.
| |
Collapse
|
17
|
Chen J, Niu C, Yang N, Liu C, Zou SS, Zhu S. Biomarker discovery and application-An opportunity to resolve the challenge of liver cancer diagnosis and treatment. Pharmacol Res 2023; 189:106674. [PMID: 36702425 DOI: 10.1016/j.phrs.2023.106674] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/10/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023]
Abstract
Liver cancer is one of the most common malignancies, with severe morbidity and mortality. While considerable progress has been made in liver cancer treatment, the 5-year overall survival (OS) of patients has not improved significantly. Reasons include the inadequate capability of early screening and diagnosis, a high incidence of recurrence and metastasis, a high degree of tumor heterogeneity, and an immunosuppressive tumor microenvironment. Therefore, the identification and validation of specific and robust liver cancer biomarkers are of major importance for early screening, timely diagnosis, accurate prognosis, and the prevention of tumor progression. In this review, we highlight some of the latest research progress and potential applications of liver cancer biomarkers, describing hotspots and prospective directions in biomarker discovery.
Collapse
Affiliation(s)
- Jingtao Chen
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, China; Laboratory for Tumor Immunology, The First Hospital of Jilin University, Changchun 130021, China
| | - Chao Niu
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Ning Yang
- Laboratory for Tumor Immunology, The First Hospital of Jilin University, Changchun 130021, China
| | - Chunyan Liu
- Laboratory for Tumor Immunology, The First Hospital of Jilin University, Changchun 130021, China
| | - Shan-Shan Zou
- Laboratory for Tumor Immunology, The First Hospital of Jilin University, Changchun 130021, China
| | - Shan Zhu
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, China; Laboratory for Tumor Immunology, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
18
|
Mossenta M, Busato D, Dal Bo M, Macor P, Toffoli G. Novel Nanotechnology Approaches to Overcome Drug Resistance in the Treatment of Hepatocellular Carcinoma: Glypican 3 as a Useful Target for Innovative Therapies. Int J Mol Sci 2022; 23:10038. [PMID: 36077433 PMCID: PMC9456072 DOI: 10.3390/ijms231710038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the second most lethal tumor, with a 5-year survival rate of 18%. Early stage HCC is potentially treatable by therapies with curative intent, whereas chemoembolization/radioembolization and systemic therapies are the only therapeutic options for intermediate or advanced HCC. Drug resistance is a critical obstacle in the treatment of HCC that could be overcome by the use of targeted nanoparticle-based therapies directed towards specific tumor-associated antigens (TAAs) to improve drug delivery. Glypican 3 (GPC3) is a member of the glypican family, heparan sulfate proteoglycans bound to the cell surface via a glycosylphosphatidylinositol anchor. The high levels of GPC3 detected in HCC and the absence or very low levels in normal and non-malignant liver make GPC3 a promising TAA candidate for targeted nanoparticle-based therapies. The use of nanoparticles conjugated with anti-GPC3 agents may improve drug delivery, leading to a reduction in severe side effects caused by chemotherapy and increased drug release at the tumor site. In this review, we describe the main clinical features of HCC and the common treatment approaches. We propose the proteoglycan GPC3 as a useful TAA for targeted therapies. Finally, we describe nanotechnology approaches for anti-GPC3 drug delivery systems based on NPs for HCC treatment.
Collapse
Affiliation(s)
- Monica Mossenta
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Davide Busato
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| |
Collapse
|
19
|
Chen H, Teng M, Zhang H, Liang X, Cheng H, Liu G. Advanced radionuclides in diagnosis and therapy for hepatocellular carcinoma. CHINESE CHEM LETT 2022; 33:3371-3383. [DOI: 10.1016/j.cclet.2022.03.079] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
20
|
Labadie KP, Hamlin DK, Kenoyer A, Daniel SK, Utria AF, Ludwig AD, Kenerson HL, Li L, Sham JG, Chen DL, Orozco JJ, Yeung RS, Orvig C, Li Y, Wilbur DS, Park JO. Glypican-3-Targeted 227Th α-Therapy Reduces Tumor Burden in an Orthotopic Xenograft Murine Model of Hepatocellular Carcinoma. J Nucl Med 2022; 63:1033-1038. [PMID: 34772791 PMCID: PMC9258570 DOI: 10.2967/jnumed.121.262562] [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: 06/03/2021] [Revised: 10/08/2021] [Indexed: 01/03/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a significant cause of morbidity and mortality worldwide, with limited therapeutic options for advanced disease. Targeted α-therapy is an emerging class of targeted cancer therapy in which α-particle-emitting radionuclides, such as 227Th, are delivered specifically to cancer tissue. Glypican-3 (GPC3) is a cell surface glycoprotein highly expressed on HCC. In this study, we describe the development and in vivo efficacy of a 227Th-labeled GPC3-targeting antibody conjugate (227Th-octapa-αGPC3) for treatment of HCC in an orthotopic murine model. Methods: The chelator p-SCN-Bn-H4octapa-NCS (octapa) was conjugated to a GPC3-targeting antibody (αGPC3) for subsequent 227Th radiolabeling (octapa-αGPC3). Conditions were varied to optimize radiolabeling of 227Th. In vitro stability was evaluated by measuring the percentage of protein-bound 227Th by γ-ray spectroscopy. An orthotopic athymic Nu/J murine model using HepG2-Red-FLuc cells was developed. Biodistribution and blood clearance of 227Th-octapa-αGPC3 were evaluated in tumor-bearing mice. The efficacy of 227Th-octapa-αGPC3 was assessed in tumor-bearing animals with serial measurement of serum α-fetoprotein at 23 d after injection. Results: Octapa-conjugated αGPC3 provided up to 70% 227Th labeling yield in 2 h at room temperature. In the presence of ascorbate, at least 97.8% of 227Th was bound to αGPC3-octapa after 14 d in phosphate-buffered saline. In HepG2-Red-FLuc tumor-bearing mice, highly specific GPC3 targeting was observed, with significant 227Th-octapa-αGPC3 accumulation in the tumor over time and minimal accumulation in normal tissue. Twenty-three days after treatment, a significant reduction in tumor burden was observed in mice receiving a 500 kBq/kg dose of 227Th-octapa-αGPC3 by tail-vein injection. No acute off-target toxicity was observed, and no animals died before termination of the study. Conclusion:227Th-octapa-αGPC3 was observed to be stable in vitro; maintain high specificity for GPC3, with favorable biodistribution in vivo; and result in significant antitumor activity without significant acute off-target toxicity in an orthotopic murine model of HCC.
Collapse
Affiliation(s)
- Kevin P. Labadie
- Department of Surgery, University of Washington, Seattle, Washington
| | - Donald K. Hamlin
- Department of Radiation Oncology, University of Washington, Seattle, Washington
| | - Aimee Kenoyer
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Sara K. Daniel
- Department of Surgery, University of Washington, Seattle, Washington
| | - Alan F. Utria
- Department of Surgery, University of Washington, Seattle, Washington
| | - Andrew D. Ludwig
- Department of Surgery, University of Washington, Seattle, Washington
| | - Heidi L. Kenerson
- Department of Surgery, University of Washington, Seattle, Washington
| | - Lily Li
- Life Sciences Division, TRIUMF, and Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jonathan G. Sham
- Department of Surgery, University of Washington, Seattle, Washington
| | - Delphine L. Chen
- Department of Radiology, University of Washington, Seattle, Washington
| | - Johnnie J. Orozco
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Raymond S. Yeung
- Department of Surgery, University of Washington, Seattle, Washington
| | - Chris Orvig
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yawen Li
- Department of Radiation Oncology, University of Washington, Seattle, Washington
| | - D. Scott Wilbur
- Department of Radiation Oncology, University of Washington, Seattle, Washington
| | - James O. Park
- Department of Surgery, University of Washington, Seattle, Washington
| |
Collapse
|
21
|
Kadassery KJ, King AP, Fayn S, Baidoo KE, MacMillan SN, Escorcia FE, Wilson JJ. H 2BZmacropa-NCS: A Bifunctional Chelator for Actinium-225 Targeted Alpha Therapy. Bioconjug Chem 2022; 33:1222-1231. [PMID: 35670495 PMCID: PMC9362842 DOI: 10.1021/acs.bioconjchem.2c00190] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Actinium-225 (225Ac) is one of the most promising radionuclides for targeted alpha therapy (TAT). With a half-life of 9.92 days and a decay chain that emits four high-energy α particles, 225Ac is well-suited for TAT when conjugated to macromolecular targeting vectors that exhibit extended in vivo circulation times. The implementation of 225Ac in these targeted constructs, however, requires a suitable chelator that can bind and retain this radionuclide in vivo. Previous work has demonstrated the suitability of a diaza-18-crown-6 macrocyclic chelator H2macropa for this application. Building upon these prior efforts, in this study, two rigid variants of H2macropa, which contain either one (H2BZmacropa) or two (H2BZ2macropa) benzene rings within the macrocyclic core, were synthesized and investigated for their potential use for 225Ac TAT. The coordination chemistry of these ligands with La3+, used as a nonradioactive model for Ac3+, was carried out. Both NMR spectroscopic and X-ray crystallographic studies of the La3+ complexes of these ligands revealed similar structural features to those found for the related complex of H2macropa. Thermodynamic stability constants of the La3+ complexes, however, were found to be 1 and 2 orders of magnitude lower than those of H2macropa for H2BZmacropa and H2BZ2macropa, respectively. The decrease in thermodynamic stability was rationalized via the use of density functional theory calculations. 225Ac radiolabeling and serum stability studies with H2BZmacropa showed that this chelator compares favorably with H2macropa. Based on these promising results, a bifunctional version of this chelator, H2BZmacropa-NCS, was synthesized and conjugated to the antibody codrituzumab (GC33), which targets the liver cancer biomarker glypican-3 (GPC3). The resulting GC33-BZmacropa conjugate and an analogous GC33-macropa conjugate were evaluated for their 225Ac radiolabeling efficiencies, antigen-binding affinities, and in vivo biodistribution in HepG2 liver cancer tumor-bearing mice. Although both conjugates were comparably effective in their radiolabeling efficiencies, [225Ac]Ac-GC33-BZmacropa showed slightly poorer serum stability and biodistribution than [225Ac]Ac-GC33-macropa. Together, these results establish H2BZmacropa-NCS as a new bifunctional chelator for the preparation of 225Ac radiopharmaceuticals.
Collapse
Affiliation(s)
- Karthika J. Kadassery
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - A. Paden King
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Stanley Fayn
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Kwamena E. Baidoo
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Samantha N. MacMillan
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Freddy E. Escorcia
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Justin J. Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
22
|
Hu A, Wilson JJ. Advancing Chelation Strategies for Large Metal Ions for Nuclear Medicine Applications. Acc Chem Res 2022; 55:904-915. [PMID: 35230803 DOI: 10.1021/acs.accounts.2c00003] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Nuclear medicine leverages radioisotopes of a wide range of elements, a significant portion of which are metals, for the diagnosis and treatment of disease. To optimally use radioisotopes of the metal ions, or radiometals, for these applications, a chelator that efficiently forms thermodynamically and kinetically stable complexes with them is required. The chelator also needs to attach to a biological targeting vector that locates pathological tissues. Numerous chelators suitable for small radiometals have been established to date, but chelators that work well for large radiometals are significantly less common. In this Account, we describe recent progress by us and others in the advancement of ligands for large radiometal chelation with emerging applications in nuclear medicine.First, we discuss and analyze the coordination chemistry of the chelator macropa, a macrocyclic ligand that contains the 18-crown-6 backbone and two picolinate pendent arms, with large metal ions in the context of nuclear medicine. This ligand is known for its unusual reverse size selectivity, the preference for binding large over small metal ions. The radiolabeling properties of macropa with large radiometals 225Ac3+, 132/135La3+, 131Ba2+, 223Ra2+, 213Bi3+, and related in vivo investigations are described. The development of macropa derivatives containing different pendent donors or rigidifying groups in the macrocyclic core is also briefly reviewed.Next, efforts to transform macropa into a radiopharmaceutical agent via covalent conjugation to biological targeting vectors are summarized. In this discussion, two types of bifunctional analogues of macropa reported in the literature, macropa-NCS and mcp-click, are presented. Their implementation in different radiopharmaceutical agents is discussed. Bioconjugates containing macropa attached to small-molecule targeting vectors or macromolecular antibodies are presented. The in vitro and in vivo evaluations of these constructs are also discussed.Lastly, chelators with dual size selectivity are described. This class of ligands exhibits good affinities for both large and small metal ions. This property is valuable for nuclear medicine applications that require the simultaneous chelation of both large and small radiometals with complementary therapeutic and diagnostic properties. Recently, we reported an 18-membered macrocyclic ligand called macrodipa that attains this selectivity pattern. This chelator, its second-generation analogue py-macrodipa, and their applications for chelating the medicinally relevant large 135La3+, 225Ac3+, 213Bi3+, and small 44Sc3+ ions are also presented. Studies with these radiometals show that py-macrodipa can effectively radiolabel and stably retain both large and small radiometals. Overall, this Account makes the case for innovative ligand design approaches for novel emerging radiometal ions with unusual coordination chemistry properties.
Collapse
Affiliation(s)
- Aohan Hu
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Justin J. Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
23
|
Hong JA, Brechbiel M, Buchsbaum J, Canaria CA, Coleman CN, Escorcia FE, Espey M, Kunos C, Lin F, Narayanan D, Capala J. National Cancer Institute support for targeted alpha-emitter therapy. Eur J Nucl Med Mol Imaging 2021; 49:64-72. [PMID: 34378064 DOI: 10.1007/s00259-021-05503-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 07/23/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Radiopharmaceutical targeted therapy (RPT) has been studied for decades; however, recent clinical trials demonstrating efficacy have helped renewed interest in the modality. METHODS This article reviews National Cancer Institute (NCI)'s support of RPT through communication via workshops and interest groups, through funding extramural programs in academia and small business, and through intramural research, including preclinical and clinical studies. RESULTS NCI has co-organized workshops and organized interest groups on RPT and RPT dosimetry to encourage the community and facilitate rigorous preclinical and clinical studies. NCI has been supporting RPT research through various mechanisms. Research has been funded through peer-reviewed NCI Research and Program Grants (RPG) and NCI Small Business Innovation Research (SBIR) Development Center, which funds small business-initiated projects, some of which have led to clinical trials. The NCI Cancer Therapy Evaluation Program (CTEP)'s Radiopharmaceutical Development Initiative supports RPT in NCI-funded clinical trials, including Imaging and Radiation Oncology Core (IROC) expertise in imaging QA and dosimetry procedures. Preclinical targeted a-emitter therapy (TAT) research at the NCI's intramural program is ongoing, building on foundational work dating back to the 1980s. Ongoing "bench-to-bedside" efforts leverage the unique infrastructure of the National Institutes of Health's (NIH) Clinical Center. CONCLUSION Given the great potential of RPT, our goal is to continue to encourage its development that will generate the high-quality evidence needed to bring this multidisciplinary treatment to patients.
Collapse
Affiliation(s)
- Julie A Hong
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr., Bethesda, MD, 20892, USA
| | - Martin Brechbiel
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Jeff Buchsbaum
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr., Bethesda, MD, 20892, USA
| | - Christie A Canaria
- Small Business Innovation Research Development Center, National Cancer Institute, Bethesda, MD, USA
| | - C Norman Coleman
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr., Bethesda, MD, 20892, USA
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Freddy E Escorcia
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD, USA
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD, USA
| | - Michael Espey
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr., Bethesda, MD, 20892, USA
| | - Charles Kunos
- Investigational Drug Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, USA
| | - Frank Lin
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD, USA
| | - Deepa Narayanan
- Small Business Innovation Research Development Center, National Cancer Institute, Bethesda, MD, USA
| | - Jacek Capala
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr., Bethesda, MD, 20892, USA.
| |
Collapse
|
24
|
Filippi L, Braat AJ. Theragnostics in primary and secondary liver tumors: the need for a personalized approach. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2021; 65:353-370. [PMID: 34881847 DOI: 10.23736/s1824-4785.21.03407-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Primary and secondary hepatic tumors have a dramatic impact in oncology. Despite many advances in diagnosis and therapy, the management of hepatic malignancies is still challenging, ranging from various loco-regional approaches to system therapies. In this scenario, theragnostic approaches, based on the administration of a radiopharmaceuticals' pair, the first labeled with a radionuclide suitable for the diagnostic phase and the second one bound to radionuclide emitting particles for therapy, is gaining more and more importance. Selective internal radiation therapy (SIRT) with microspheres labeled with 90Y or 166Ho is widely used as a loco-regional treatment for primary and secondary hepatic tumors. While 166Ho presents both gamma and beta emission and can be therefore considered a real "theragnostic" agent, for 90Y-microspheres theragnostic approach is realized at the diagnostic phase through the utilization of macroaggregates of human albumin, labeled with 99mTc as "biosimilar" agent respect to microspheres. The aim of the present review was to cover theragnostic applications of 90Y/166Ho-labeled microspheres in clinical practice. Furthermore, we report the preliminary data concerning the potential role of some emerging theragnostic biomarkers for hepatocellular carcinoma, such as glypican-3 (GPC3) and prostate specific membrane antigen (PSMA).
Collapse
Affiliation(s)
- Luca Filippi
- Department of Nuclear Medicine, Santa Maria Goretti Hospital, Latina, Italy -
| | - Arthur J Braat
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
25
|
Eychenne R, Chérel M, Haddad F, Guérard F, Gestin JF. Overview of the Most Promising Radionuclides for Targeted Alpha Therapy: The "Hopeful Eight". Pharmaceutics 2021; 13:pharmaceutics13060906. [PMID: 34207408 PMCID: PMC8234975 DOI: 10.3390/pharmaceutics13060906] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 12/11/2022] Open
Abstract
Among all existing radionuclides, only a few are of interest for therapeutic applications and more specifically for targeted alpha therapy (TAT). From this selection, actinium-225, astatine-211, bismuth-212, bismuth-213, lead-212, radium-223, terbium-149 and thorium-227 are considered as the most suitable. Despite common general features, they all have their own physical characteristics that make them singular and so promising for TAT. These radionuclides were largely studied over the last two decades, leading to a better knowledge of their production process and chemical behavior, allowing for an increasing number of biological evaluations. The aim of this review is to summarize the main properties of these eight chosen radionuclides. An overview from their availability to the resulting clinical studies, by way of chemical design and preclinical studies is discussed.
Collapse
Affiliation(s)
- Romain Eychenne
- Groupement d’Intérêt Public ARRONAX, 1 Rue Aronnax, F-44817 Saint-Herblain, France;
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Cancérologie et Immunologie Nantes—Angers (CRCINA)—UMR 1232, ERL 6001, F-44000 Nantes, France; (M.C.); (F.G.)
- Correspondence: (R.E.); (J.-F.G.)
| | - Michel Chérel
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Cancérologie et Immunologie Nantes—Angers (CRCINA)—UMR 1232, ERL 6001, F-44000 Nantes, France; (M.C.); (F.G.)
| | - Férid Haddad
- Groupement d’Intérêt Public ARRONAX, 1 Rue Aronnax, F-44817 Saint-Herblain, France;
- Laboratoire Subatech, UMR 6457, Université de Nantes, IMT Atlantique, CNRS, Subatech, F-44000 Nantes, France
| | - François Guérard
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Cancérologie et Immunologie Nantes—Angers (CRCINA)—UMR 1232, ERL 6001, F-44000 Nantes, France; (M.C.); (F.G.)
| | - Jean-François Gestin
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Cancérologie et Immunologie Nantes—Angers (CRCINA)—UMR 1232, ERL 6001, F-44000 Nantes, France; (M.C.); (F.G.)
- Correspondence: (R.E.); (J.-F.G.)
| |
Collapse
|
26
|
Singh A, Beechinor RJ, Huynh JC, Li D, Dayyani F, Valerin JB, Hendifar A, Gong J, Cho M. Immunotherapy Updates in Advanced Hepatocellular Carcinoma. Cancers (Basel) 2021; 13:cancers13092164. [PMID: 33946408 PMCID: PMC8125389 DOI: 10.3390/cancers13092164] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Advanced hepatocellular carcinoma (HCC) carries a grim prognosis, which has historically been compounded by a lack of available systemic therapies. Sorafenib monotherapy was the standard of care for front-line treatment of advanced HCC for many years, despite both poor tolerability and lack of durable responses. In the past few years, there have been several clinical trials evaluating the efficacy of immune checkpoint inhibitors for advanced HCC. Use of immune checkpoint inhibitors alone, and in combination with targeted therapies, has led to improved outcomes in both treatment-naïve and subsequent line treatment of advanced HCC. Here we review the role of immunotherapy in the treatment of HCC, describe the mechanistic basis for combination with targeted therapy, and summarize the recent published data as well as ongoing clinical trials for the use of immunotherapy in the treatment of advanced HCC. Abstract Hepatocellular carcinoma (HCC) is the second most common cause of cancer death worldwide. HCC tumor development and treatment resistance are impacted by changes in the microenvironment of the hepatic immune system. Immunotherapy has the potential to improve response rates by overcoming immune tolerance mechanisms and strengthening anti-tumor activity in the tumor microenvironment. In this review, we characterize the impact of immunotherapy on outcomes of advanced HCC, as well as the active clinical trials evaluating novel combination immunotherapy strategies. In particular, we discuss the efficacy of atezolizumab and bevacizumab as demonstrated in the IMbrave150 study, which created a new standard of care for the front-line treatment of advanced HCC. However, there are multiple ongoing trials that may present additional front-line treatment options depending on their efficacy/toxicity results. Furthermore, the preliminary data on the application of chimeric antigen receptor (CAR-T) cell therapy for treatment of HCC suggests this may be a promising option for the future of advanced HCC treatment.
Collapse
Affiliation(s)
- Amisha Singh
- Internal Medicine, University of California, Davis, Sacramento, CA 95817, USA;
| | | | - Jasmine C. Huynh
- Hematology Oncology, University of California, Davis, Sacramento, CA 95817, USA;
| | - Daneng Li
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA 91010, USA;
| | - Farshid Dayyani
- Hematology Oncology, University of California, Irvine, Irvine, CA 92868, USA; (F.D.); (J.B.V.)
| | - Jennifer B. Valerin
- Hematology Oncology, University of California, Irvine, Irvine, CA 92868, USA; (F.D.); (J.B.V.)
| | - Andrew Hendifar
- Hematology Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.H.); (J.G.)
| | - Jun Gong
- Hematology Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.H.); (J.G.)
| | - May Cho
- Hematology Oncology, University of California, Irvine, Irvine, CA 92868, USA; (F.D.); (J.B.V.)
- Correspondence:
| |
Collapse
|
27
|
White JM, Escorcia FE, Viola NT. Perspectives on metals-based radioimmunotherapy (RIT): moving forward. Theranostics 2021; 11:6293-6314. [PMID: 33995659 PMCID: PMC8120204 DOI: 10.7150/thno.57177] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/22/2021] [Indexed: 12/18/2022] Open
Abstract
Radioimmunotherapy (RIT) is FDA-approved for the clinical management of liquid malignancies, however, its use for solid malignancies remains a challenge. The putative benefit of RIT lies in selective targeting of antigens expressed on the tumor surface using monoclonal antibodies, to systemically deliver cytotoxic radionuclides. The past several decades yielded dramatic improvements in the quality, quantity, recent commercial availability of alpha-, beta- and Auger Electron-emitting therapeutic radiometals. Investigators have created new or improved existing bifunctional chelators. These bifunctional chelators bind radiometals and can be coupled to antigen-specific antibodies. In this review, we discuss approaches to develop radiometal-based RITs, including the selection of radiometals, chelators and antibody platforms (i.e. full-length, F(ab')2, Fab, minibodies, diabodies, scFv-Fc and nanobodies). We cite examples of the performance of RIT in the clinic, describe challenges to its implementation, and offer insights to address gaps toward translation.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/therapeutic use
- Antigens, Neoplasm/immunology
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/metabolism
- Antineoplastic Agents, Immunological/therapeutic use
- Chelating Agents/administration & dosage
- Chelating Agents/metabolism
- Click Chemistry
- Clinical Trials as Topic
- Dose Fractionation, Radiation
- Drug Delivery Systems
- Forecasting
- Humans
- Immunoglobulin Fab Fragments/administration & dosage
- Immunoglobulin Fab Fragments/therapeutic use
- Lymphoma, Non-Hodgkin/radiotherapy
- Mice
- Molecular Targeted Therapy
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasms, Experimental/diagnostic imaging
- Neoplasms, Experimental/radiotherapy
- Organ Specificity
- Precision Medicine
- Radiation Tolerance
- Radioimmunotherapy/methods
- Radiopharmaceuticals/administration & dosage
- Radiopharmaceuticals/therapeutic use
- Receptor Protein-Tyrosine Kinases/antagonists & inhibitors
- Single-Chain Antibodies/administration & dosage
- Single-Chain Antibodies/therapeutic use
- Single-Domain Antibodies/administration & dosage
- Single-Domain Antibodies/therapeutic use
- Yttrium Radioisotopes/administration & dosage
- Yttrium Radioisotopes/therapeutic use
Collapse
Affiliation(s)
- Jordan M. White
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201
- Department of Oncology, Karmanos Cancer Institute, Detroit, MI 48201
| | - Freddy E. Escorcia
- Molecular Imaging Branch, Radiation Oncology Branch, National Cancer Institute, Bethesda, MD 20814
| | - Nerissa T. Viola
- Department of Oncology, Karmanos Cancer Institute, Detroit, MI 48201
| |
Collapse
|