1
|
Veire JL, Lucas MJ, Bond LG, Tripu DR, Tessier PM, Greineder CF. Comparative Radiotracing Quantifies Brain Cellular Uptake and Catabolism of Bispecific Antibodies Targeting Transferrin Receptor and CD98hc. ACS Chem Neurosci 2025; 16:1264-1274. [PMID: 40071777 DOI: 10.1021/acschemneuro.4c00552] [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] [Indexed: 04/03/2025] Open
Abstract
Bispecific antibodies (bAbs) that engage cerebrovascular targets, induce transport across the blood-brain barrier (BBB), and redistribute to secondary targets within the brain parenchyma have the potential to transform the diagnosis and treatment of a wide range of central nervous system disorders. Full understanding of the pharmacokinetics (PK) of these agents, including their potential for delivering cargo into brain parenchymal cells, is a key priority for the development of numerous potential therapeutic applications. To date, the brain PK of bAbs that target transferrin receptor (TfR-1) and CD98 heavy chain (CD98hc) has been characterized using techniques incapable of distinguishing between CNS clearance of intact protein from uptake and catabolism by brain parenchymal cells. Herein, we address this knowledge gap via a comparative radiotracing strategy using two radioisotopes with distinct residualizing properties, iodine-125 (I-125) and zirconium-89 (Zr-89). We first identify reaction conditions for tetravalent chelator modification and Zr-89 radiolabeling that do not adversely affect in vitro or in vivo function. We then use comparative radiotracing to define the PK of TfR-1 and CD98hc targeted bAbs without a parenchymal target, generating quantitative evidence of TfR-1-mediated cellular uptake and catabolism that implicates these processes in previously reported differences in the brain retention of IgGs shuttled across the BBB via these two pathways. Finally, we perform comparative radiotracing on a TfR-1 bAb with an internalizing neuronal target (TrkB), demonstrating rapid divergence of Zr-89 and I-125 PK curves, with a > 30-fold difference in brain content of the two radioisotopes. Together, these results establish comparative radiotracing as a valuable technique for identifying internalizing cellular targets within the brain parenchyma and quantifying the extent and timing of bAb uptake and catabolism following target engagement.
Collapse
Affiliation(s)
- Jacob L Veire
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Michael J Lucas
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Layne G Bond
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Deepika R Tripu
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Peter M Tessier
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Colin F Greineder
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Emergency Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
2
|
Cho N, Ho J, Rosario GD, Yu SF, Ferl GZ, Boswell CA. Measurement of specific and nonspecific tissue uptake of antibodies in tumor by SPECT imaging and nonlinear compartmental modeling. EJNMMI Res 2025; 15:15. [PMID: 39994098 PMCID: PMC11850665 DOI: 10.1186/s13550-025-01207-9] [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: 12/12/2024] [Accepted: 02/14/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Understanding the mechanisms driving specific and nonspecific tissue uptake of antibodies can inform protein engineering strategies that maximize therapeutic efficacy in target tissues while minimizing off-target tissue toxicities. While in vitro cell assays are typically used to study these internalization mechanisms, there are few methods readily available to evaluate these pathways in vivo. Single photon emission computed tomography (SPECT) imaging with a non-residualizing radiohalogen probe can measure total levels of intact antibody, and a residualizing radiometal-chelate probe, in combination with a non-residualizing probe, can measure catabolized antibody associated with receptor-mediated and nonspecific internalization processes. Here, we describe a SPECT imaging study in human epidermal growth factor receptor 2 (HER2)-expressing tumor-bearing mice aimed at measuring whole body disposition kinetics of tumor-targeting trastuzumab (anti-HER2) and non-targeting (anti-gD) antibodies. Mice received these molecules labeled with either a non-residualizing prosthetic group ([125I]SIB) or with a residualizing radiometal-chelate (111In-DOTA). RESULTS SPECT imaging data confirmed significant HER2-mediated tumor uptake and catabolism of anti-HER2, evidenced by the high 111In-DOTA-anti-HER2 signal over time relative to 111In-DOTA-anti-gD and the respective [125I]SIB-labeled molecules. [125I]SIB-anti-HER2 still showed noticeably higher tumor signal than [125I]SIB-anti-gD, demonstrating a meaningful pool of intact anti-HER2 in the interstitial tumor compartment. Spleen showed the greatest catabolism of both mAbs amongst all non-tumor tissues. Compartmental modeling of the SPECT data demonstrated that cell-associated anti-HER2 was primarily receptor-bound, with a peak receptor occupancy of 35% at 13 h post administration of a 10 mg/kg dose, with minimal free and pinocytosed mAb. CONCLUSION Here, we successfully developed an imaging and modeling approach to capture anti-HER2 antibody receptor binding as well as specific and nonspecific internalization over time in vivo. These data and analyses demonstrate the power of SPECT imaging using both non-residualizing and residualizing radioisotopes to better characterize the different biological states (free, bound, and catabolized) of antibodies within interstitial and intracellular compartments. Understanding these distinct antibody internalization mechanisms in tumor and non-tumor tissues enables more informed decisions on dose selection to optimize treatment of tumors with heterogeneous antigen expression while minimizing nonspecific toxicities.
Collapse
Affiliation(s)
- Nicholas Cho
- Department of Preclinical and Translational Pharmacokinetics, Genentech Research and Early Development, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Jason Ho
- In Vivo Studies, Genentech Research and Early Development, South San Francisco, CA, 94080, USA
| | - Geoffrey Del Rosario
- Translational Oncology, Genentech Research and Early Development, South San Francisco, CA, 94080, USA
| | - Shang-Fan Yu
- Translational Oncology, Genentech Research and Early Development, South San Francisco, CA, 94080, USA
| | - Gregory Z Ferl
- Department of Preclinical and Translational Pharmacokinetics, Genentech Research and Early Development, 1 DNA Way, South San Francisco, CA, 94080, USA.
- Translational Imaging, Genentech Research and Early Development, South San Francisco, CA, 94080, USA.
| | - C Andrew Boswell
- Department of Preclinical and Translational Pharmacokinetics, Genentech Research and Early Development, 1 DNA Way, South San Francisco, CA, 94080, USA.
- Translational Imaging, Genentech Research and Early Development, South San Francisco, CA, 94080, USA.
| |
Collapse
|
3
|
Garaulet G, Báez BB, Medrano G, Rivas-Sánchez M, Sánchez-Alonso D, Martinez-Torrecuadrada JL, Mulero F. Radioimmunotheragnosis in Cancer Research. Cancers (Basel) 2024; 16:2896. [PMID: 39199666 PMCID: PMC11352548 DOI: 10.3390/cancers16162896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
The combination of immunoPET-where an antibody (Ab) is labeled with an isotope for PET imaging-and radioimmunotherapy (RIT), using the same antibody with a therapeutic isotope, offers significant advantages in cancer management. ImmunoPET allows non-invasive imaging of antigen expression, which aids in patient selection for subsequent radioimmunotherapy. It also facilitates the assessment of tumor response to therapy, allowing for treatment adjustments if necessary. In addition, immunoPET provides critical pharmacokinetic data, including antibody biodistribution and clearance rates, which are essential for dosimetry calculations and treatment protocol optimization. There are still challenges to overcome. Identifying appropriate target antigens that are selectively expressed on cancer cells while minimally expressed on normal tissues remains a major hurdle to reduce off-target toxicity. In addition, it is critical to optimize the pharmacokinetics of radiolabeled antibodies to maximize tumor uptake and minimize normal tissue uptake, particularly in vital organs such as the liver and kidney. This approach offers the potential for targeted and personalized cancer therapy with reduced systemic toxicity by exploiting the specificity of monoclonal antibodies and the cytotoxic effects of radiation. However, further research is needed to address remaining challenges and to optimize these technologies for clinical use.
Collapse
Affiliation(s)
- Guillermo Garaulet
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| | - Bárbara Beatriz Báez
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| | - Guillermo Medrano
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| | - María Rivas-Sánchez
- Protein Production Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (M.R.-S.); (D.S.-A.)
| | - David Sánchez-Alonso
- Protein Production Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (M.R.-S.); (D.S.-A.)
| | | | - Francisca Mulero
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| |
Collapse
|
4
|
Mohr P, van Sluis J, Lub-de Hooge MN, Lammertsma AA, Brouwers AH, Tsoumpas C. Advances and challenges in immunoPET methodology. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 4:1360710. [PMID: 39355220 PMCID: PMC11440922 DOI: 10.3389/fnume.2024.1360710] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 02/05/2024] [Indexed: 10/03/2024]
Abstract
Immuno-positron emission tomography (immunoPET) enables imaging of specific targets that play a role in targeted therapy and immunotherapy, such as antigens on cell membranes, targets in the disease microenvironment, or immune cells. The most common immunoPET applications use a monoclonal antibody labeled with a relatively long-lived positron emitter such as 89Zr (T 1/2 = 78.4 h), but smaller antibody-based constructs labeled with various other positron emitting radionuclides are also being investigated. This molecular imaging technique can thus guide the development of new drugs and may have a pivotal role in selecting patients for a particular therapy. In early phase immunoPET trials, multiple imaging time points are used to examine the time-dependent biodistribution and to determine the optimal imaging time point, which may be several days after tracer injection due to the slow kinetics of larger molecules. Once this has been established, usually only one static scan is performed and semi-quantitative values are reported. However, total PET uptake of a tracer is the sum of specific and nonspecific uptake. In addition, uptake may be affected by other factors such as perfusion, pre-/co-administration of the unlabeled molecule, and the treatment schedule. This article reviews imaging methodologies used in immunoPET studies and is divided into two parts. The first part summarizes the vast majority of clinical immunoPET studies applying semi-quantitative methodologies. The second part focuses on a handful of studies applying pharmacokinetic models and includes preclinical and simulation studies. Finally, the potential and challenges of immunoPET quantification methodologies are discussed within the context of the recent technological advancements provided by long axial field of view PET/CT scanners.
Collapse
Affiliation(s)
- Philipp Mohr
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Joyce van Sluis
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marjolijn N Lub-de Hooge
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adriaan A Lammertsma
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adrienne H Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Charalampos Tsoumpas
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
5
|
Zhang H, Xiao L, Xie H, Li L. Hotspots and frontiers in PSMA research for prostate cancer: a bibliometric and visualization analysis over the past 20 years. Eur J Med Res 2023; 28:610. [PMID: 38115121 PMCID: PMC10731714 DOI: 10.1186/s40001-023-01590-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/12/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Prostate-specific membrane antigen (PSMA)-targeted imaging and therapy have significantly changed the management of patients with prostate cancer (PCa) at different disease stages. This advancement has attracted the attention of scholars, leading to a prolific output of scholarly publications. This study comprehensively outlines the knowledge framework associated with PSMA-based diagnosis and treatment of PCa through the application of bibliometric analysis, and discusses the potential research trends and foci. METHODS Articles and reviews related to PSMA for prostate cancer from 2003 to 2022 were retrieved from Web of Science Core Collection. VOSviewer, Citespace, and R-bibliometrix were primarily employed to execute and visually represent co-authorship, co-citation, and co-occurrence analysis of countries, institutions, authors, references and keywords in this field. RESULTS A total of 3830 papers were included. The papers on the field of PSMA-based PCa therapy and imaging had been continuously increased since 2003, but the rate has slowed from 2020. The United States made the largest contribution in this field, in terms of publications 997 (26.03%), H-index (110) and total citations (53,167 times). We identified the most productive institution were Technical University of Munich, and Australian institutions had become very active in recent years. Journal of Nuclear Medicine was the most prominent journal in this field. Professors Matthias Eiber and Martin G Pomper made great achievements, while Ali Afshar-Oromieh was the most co-cited author. According to the result of keywords and topics analysis, "ga-68 labeled psma ligand", "radiation dosimetry" and "HBED-CC" were major research areas in the near future, while "Extended pelvic lymph node dissection" was considered to be the future research foci. CONCLUSIONS The field of psma-based PCa therapy and imaging is in the stage of vigorous development and has a bright prospect. The United States and Germany have achieved outstanding results in this area, while Australia has recently developed rapidly. It is foreseeable that more research foci will be lied in the early detection of pelvic lymph nodes and the multimodal imaging-guided surgery.
Collapse
Affiliation(s)
- Hanfei Zhang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, 610041, China
| | - Liu Xiao
- Department of Nuclear Medicine, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, 610041, China
| | - Hangyu Xie
- Department of Nuclear Medicine, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, 610041, China
| | - Lin Li
- Department of Nuclear Medicine, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, 610041, China.
| |
Collapse
|
6
|
Accommodation in allogeneic and xenogeneic organ transplantation: Prevalence, impact, and implications for monitoring and for therapeutics. Hum Immunol 2023; 84:5-17. [PMID: 36244871 DOI: 10.1016/j.humimm.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/01/2022] [Indexed: 11/04/2022]
Abstract
Accommodation refers to acquired resistance of organs or tissues to immune or inflammatory reactions that might otherwise cause severe injury or rejection. As first observed in ABO-incompatible kidney transplants and heterotopic cardiac xenografts, accommodation was identified when organ transplants continued to function despite the presence of anti-graft antibodies and/or other reactants in the blood of recipients. Recent evidence suggests many and perhaps most organ transplants have accommodation, as most recipients mount B cell responses specific for the graft. Wide interest in the impact of graft-specific antibodies on the outcomes of transplants prompts questions about which mechanisms confer protection against such antibodies, how accommodation might be detected and whether and how rejection could be superimposed on accommodation. Xenotransplantation offers a unique opportunity to address these questions because immune responses to xenografts are easily detected and the pathogenic impact of immune responses is so severe. Xenotransplantation also provides a compelling need to apply these and other insights to decrease the intensity and toxicity of immunosuppression that otherwise could limit clinical application.
Collapse
|
7
|
Lugat A, Bailly C, Chérel M, Rousseau C, Kraeber-Bodéré F, Bodet-Milin C, Bourgeois M. Immuno-PET: Design options and clinical proof-of-concept. Front Med (Lausanne) 2022; 9:1026083. [PMID: 36314010 PMCID: PMC9613928 DOI: 10.3389/fmed.2022.1026083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/29/2022] [Indexed: 11/23/2022] Open
Abstract
Radioimmunoconjugates have been used for over 30 years in nuclear medicine applications. In the last few years, advances in cancer biology knowledge have led to the identification of new molecular targets specific to certain patient subgroups. The use of these targets in targeted therapies approaches has allowed the developments of specifically tailored therapeutics for patients. As consequence of the PET-imaging progresses, nuclear medicine has developed powerful imaging tools, based on monoclonal antibodies, to in vivo characterization of these tumor biomarkers. This imaging modality known as immuno-positron emission tomography (immuno-PET) is currently in fastest-growing and its medical value lies in its ability to give a non-invasive method to assess the in vivo target expression and distribution and provide key-information on the tumor targeting. Currently, immuno-PET presents promising probes for different nuclear medicine topics as staging/stratification tool, theranostic approaches or predictive/prognostic biomarkers. To develop a radiopharmaceutical drug that can be used in immuno-PET approach, it is necessary to find the best compromise between the isotope choice and the immunologic structure (full monoclonal antibody or derivatives). Through some clinical applications, this paper review aims to discuss the most important aspects of the isotope choice and the usable proteic structure that can be used to meet the clinical needs.
Collapse
Affiliation(s)
- Alexandre Lugat
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France
| | - Clément Bailly
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France
| | - Michel Chérel
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Department of Nuclear Medicine, Institut de Cancérologie de l'Ouest (ICO) – Site Gauducheau, Saint-Herblain, France
| | - Caroline Rousseau
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Department of Nuclear Medicine, Institut de Cancérologie de l'Ouest (ICO) – Site Gauducheau, Saint-Herblain, France
| | - Françoise Kraeber-Bodéré
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France
| | - Caroline Bodet-Milin
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France
| | - Mickaël Bourgeois
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France,ARRONAX Cyclotron, Saint-Herblain, France,*Correspondence: Mickaël Bourgeois
| |
Collapse
|
8
|
Johnson RP, Ratnacaram CK, Kumar L, Jose J. Combinatorial approaches of nanotherapeutics for inflammatory pathway targeted therapy of prostate cancer. Drug Resist Updat 2022; 64:100865. [PMID: 36099796 DOI: 10.1016/j.drup.2022.100865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 12/24/2022]
Abstract
Prostate cancer (PC) is the most prevalent male urogenital cancer worldwide. PC patients presenting an advanced or metastatic cancer succumb to the disease, even after therapeutic interventions including radiotherapy, surgery, androgen deprivation therapy (ADT), and chemotherapy. One of the hallmarks of PC is evading immune surveillance and chronic inflammation, which is a major challenge towards designing effective therapeutic formulations against PC. Chronic inflammation in PC is often characterized by tumor microenvironment alterations, epithelial-mesenchymal transition and extracellular matrix modifications. The inflammatory events are modulated by reactive nitrogen and oxygen species, inflammatory cytokines and chemokines. Major signaling pathways in PC includes androgen receptor, PI3K and NF-κB pathways and targeting these inter-linked pathways poses a major therapeutic challenge. Notably, many conventional treatments are clinically unsuccessful, due to lack of targetability and poor bioavailability of the therapeutics, untoward toxicity and multidrug resistance. The past decade witnessed an advancement of nanotechnology as an excellent therapeutic paradigm for PC therapy. Modern nanovectorization strategies such as stimuli-responsive and active PC targeting carriers offer controlled release patterns and superior anti-cancer effects. The current review initially describes the classification, inflammatory triggers and major inflammatory pathways of PC, various PC treatment strategies and their limitations. Subsequently, recent advancement in combinatorial nanotherapeutic approaches, which target PC inflammatory pathways, and the mechanism of action are discussed. Besides, the current clinical status and prospects of PC homing nanovectorization, and major challenges to be addressed towards the advancement PC therapy are also addressed.
Collapse
Affiliation(s)
- Renjith P Johnson
- Polymer Nanobiomaterial Research Laboratory, Nanoscience and Microfluidics Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Chandrahas Koumar Ratnacaram
- Cell Signaling and Cancer Biology Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Lalit Kumar
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka 576 104, India
| | - Jobin Jose
- NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Mangalore 575018, India.
| |
Collapse
|
9
|
Manafi-Farid R, Ataeinia B, Ranjbar S, Jamshidi Araghi Z, Moradi MM, Pirich C, Beheshti M. ImmunoPET: Antibody-Based PET Imaging in Solid Tumors. Front Med (Lausanne) 2022; 9:916693. [PMID: 35836956 PMCID: PMC9273828 DOI: 10.3389/fmed.2022.916693] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/24/2022] [Indexed: 12/13/2022] Open
Abstract
Immuno-positron emission tomography (immunoPET) is a molecular imaging modality combining the high sensitivity of PET with the specific targeting ability of monoclonal antibodies. Various radioimmunotracers have been successfully developed to target a broad spectrum of molecules expressed by malignant cells or tumor microenvironments. Only a few are translated into clinical studies and barely into clinical practices. Some drawbacks include slow radioimmunotracer kinetics, high physiologic uptake in lymphoid organs, and heterogeneous activity in tumoral lesions. Measures are taken to overcome the disadvantages, and new tracers are being developed. In this review, we aim to mention the fundamental components of immunoPET imaging, explore the groundbreaking success achieved using this new technique, and review different radioimmunotracers employed in various solid tumors to elaborate on this relatively new imaging modality.
Collapse
Affiliation(s)
- Reyhaneh Manafi-Farid
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahar Ataeinia
- Department of Radiology, Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Shaghayegh Ranjbar
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Zahra Jamshidi Araghi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mobin Moradi
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Christian Pirich
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Mohsen Beheshti
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
10
|
Crișan G, Moldovean-Cioroianu NS, Timaru DG, Andrieș G, Căinap C, Chiș V. Radiopharmaceuticals for PET and SPECT Imaging: A Literature Review over the Last Decade. Int J Mol Sci 2022; 23:5023. [PMID: 35563414 PMCID: PMC9103893 DOI: 10.3390/ijms23095023] [Citation(s) in RCA: 128] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/23/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Positron emission tomography (PET) uses radioactive tracers and enables the functional imaging of several metabolic processes, blood flow measurements, regional chemical composition, and/or chemical absorption. Depending on the targeted processes within the living organism, different tracers are used for various medical conditions, such as cancer, particular brain pathologies, cardiac events, and bone lesions, where the most commonly used tracers are radiolabeled with 18F (e.g., [18F]-FDG and NA [18F]). Oxygen-15 isotope is mostly involved in blood flow measurements, whereas a wide array of 11C-based compounds have also been developed for neuronal disorders according to the affected neuroreceptors, prostate cancer, and lung carcinomas. In contrast, the single-photon emission computed tomography (SPECT) technique uses gamma-emitting radioisotopes and can be used to diagnose strokes, seizures, bone illnesses, and infections by gauging the blood flow and radio distribution within tissues and organs. The radioisotopes typically used in SPECT imaging are iodine-123, technetium-99m, xenon-133, thallium-201, and indium-111. This systematic review article aims to clarify and disseminate the available scientific literature focused on PET/SPECT radiotracers and to provide an overview of the conducted research within the past decade, with an additional focus on the novel radiopharmaceuticals developed for medical imaging.
Collapse
Affiliation(s)
- George Crișan
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
- Department of Nuclear Medicine, County Clinical Hospital, Clinicilor 3-5, 400006 Cluj-Napoca, Romania;
| | | | - Diana-Gabriela Timaru
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
| | - Gabriel Andrieș
- Department of Nuclear Medicine, County Clinical Hospital, Clinicilor 3-5, 400006 Cluj-Napoca, Romania;
| | - Călin Căinap
- The Oncology Institute “Prof. Dr. Ion Chiricuţă”, Republicii 34-36, 400015 Cluj-Napoca, Romania;
| | - Vasile Chiș
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
- Institute for Research, Development and Innovation in Applied Natural Sciences, Babeș-Bolyai University, Str. Fântânele 30, 400327 Cluj-Napoca, Romania
| |
Collapse
|
11
|
Kiraga Ł, Kucharzewska P, Paisey S, Cheda Ł, Domańska A, Rogulski Z, Rygiel TP, Boffi A, Król M. Nuclear imaging for immune cell tracking in vivo – Comparison of various cell labeling methods and their application. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
12
|
Vázquez SM, Endepols H, Fischer T, Tawadros SG, Hohberg M, Zimmermanns B, Dietlein F, Neumaier B, Drzezga A, Dietlein M, Schomäcker K. Translational Development of a Zr-89-Labeled Inhibitor of Prostate-specific Membrane Antigen for PET Imaging in Prostate Cancer. Mol Imaging Biol 2021; 24:115-125. [PMID: 34370181 PMCID: PMC8760230 DOI: 10.1007/s11307-021-01632-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/06/2021] [Accepted: 07/12/2021] [Indexed: 12/26/2022]
Abstract
Purpose We present here a Zr-89-labeled inhibitor of prostate-specific membrane antigen (PSMA) as a complement to the already established F-18- or Ga-68-ligands. Procedures The precursor PSMA-DFO (ABX) was used for Zr-89-labeling. This is not an antibody, but a peptide analogue of the precursor for the production of [177Lu]Lu-PSMA-617. The ligand [89Zr]Zr-PSMA-DFO was compared with [68Ga]Ga-PSMA-11 and [18F]F-JK-PSMA-7 in vitro by determination of the Kd value, cellular uptake, internalization in LNCaP cells, biodistribution studies with LNCaP prostate tumor xenografts in mice, and in vivo by small-animal PET imaging in LNCaP tumor mouse models. A first-in-human PET was performed with [89Zr]Zr-PSMA-DFO on a patient presenting with a biochemical recurrence after brachytherapy and an ambiguous intraprostatic finding with [18F]F-JK-PSMA-7 but histologically benign cells in a prostate biopsy 7 months previously. Results [89Zr]Zr-PSMA-DFO was prepared with a radiochemical purity ≥ 99.9% and a very high in vitro stability for up to 7 days at 37 °C. All radiotracers showed similar specific cellular binding and internalization, in vitro and comparable tumor uptake in biodistribution experiments during the first 5 h. The [89Zr]Zr-PSMA-DFO achieved significantly higher tumor/background ratios in LNCaP tumor xenografts (tumor/blood: 309 ± 89, tumor/muscle: 450 ± 38) after 24 h than [68Ga]Ga-PSMA-11 (tumor/blood: 112 ± 57, tumor/muscle: 58 ± 36) or [18F]F-JK-PSMA-7 (tumor/blood: 175 ± 30, tumor/muscle: 114 ± 14) after 4 h (p < 0.01). Small-animal PET imaging demonstrated in vivo that tumor visualization with [89Zr]Zr-PSMA-DFO is comparable to [68Ga]Ga-PSMA-11 or [18F]F-JK-PSMA-7 at early time points (1 h p.i.) and that PET scans up to 48 h p.i. clearly visualized the tumor at late time points. A late [89Zr]Zr-PSMA-DFO PET scan on a patient with biochemical recurrence (BCR) had demonstrated intensive tracer accumulation in the right (SUVmax 13.25, 48 h p.i.) and in the left prostate lobe (SUV max 9.47), a repeat biopsy revealed cancer cells on both sides. Conclusion [89Zr]Zr-PSMA-DFO is a promising PSMA PET tracer for detection of tumor areas with lower PSMA expression and thus warrants further clinical evaluation. Supplementary Information The online version contains supplementary material available at 10.1007/s11307-021-01632-x.
Collapse
Affiliation(s)
- Sergio Muñoz Vázquez
- Faculty of Medicine and University Hospital Cologne, Department of Nuclear Medicine, University of Cologne, Kerpener Str. 62 50937, Cologne, Germany
| | - Heike Endepols
- Faculty of Medicine and University Hospital Cologne, Department of Nuclear Medicine, University of Cologne, Kerpener Str. 62 50937, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Kerpener Str. 62 50937, Cologne, Germany.,Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Straße 52428, Jülich, Germany
| | - Thomas Fischer
- Faculty of Medicine and University Hospital Cologne, Department of Nuclear Medicine, University of Cologne, Kerpener Str. 62 50937, Cologne, Germany
| | - Samir-Ghali Tawadros
- Faculty of Medicine and University Hospital Cologne, Center for Experimental Medicine (CEM), University of Cologne, Robert-Koch-Straße 10 50931, Cologne, Germany
| | - Melanie Hohberg
- Faculty of Medicine and University Hospital Cologne, Department of Nuclear Medicine, University of Cologne, Kerpener Str. 62 50937, Cologne, Germany
| | - Beate Zimmermanns
- Faculty of Medicine and University Hospital Cologne, Department of Nuclear Medicine, University of Cologne, Kerpener Str. 62 50937, Cologne, Germany
| | - Felix Dietlein
- Faculty of Medicine and University Hospital Cologne, Department of Nuclear Medicine, University of Cologne, Kerpener Str. 62 50937, Cologne, Germany.,Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Bernd Neumaier
- Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Kerpener Str. 62 50937, Cologne, Germany.,Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Straße 52428, Jülich, Germany
| | - Alexander Drzezga
- Faculty of Medicine and University Hospital Cologne, Department of Nuclear Medicine, University of Cologne, Kerpener Str. 62 50937, Cologne, Germany
| | - Markus Dietlein
- Faculty of Medicine and University Hospital Cologne, Department of Nuclear Medicine, University of Cologne, Kerpener Str. 62 50937, Cologne, Germany
| | - Klaus Schomäcker
- Faculty of Medicine and University Hospital Cologne, Department of Nuclear Medicine, University of Cologne, Kerpener Str. 62 50937, Cologne, Germany.
| |
Collapse
|
13
|
ANO7: Insights into topology, function, and potential applications as a biomarker and immunotherapy target. Tissue Cell 2021; 72:101546. [PMID: 33940566 DOI: 10.1016/j.tice.2021.101546] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 03/21/2021] [Accepted: 04/11/2021] [Indexed: 01/01/2023]
Abstract
Anoctamin 7 (ANO7) is a member of the transmembrane protein TMEM16 family. It has a conservative topology similar to other members in this family, such as the typical eight-transmembrane domain, but it also has unique features. Although the ion channel role of ANO7 has been well accepted, evolutionary analyses and relevant studies suggest that ANO7 may be a multi-facet protein in function. Studies have shown that ANO7 may also function as a scramblase. ANO7 is highly expressed in prostate cancer as well as normal prostate tissues. A considerable amount of evidence has confirmed that ANO7 is associated with human physiology and pathology, particularly with the development of prostate cancer, which makes ANO7 a good candidate as a diagnostic and prognostic biomarker. In addition, ANO7 may be a potential target for prostate cancer immunotherapy. Antibody-based or T cell-mediated immunotherapies against prostate cancer by targeting ANO7 have been highly anticipated. ANO7 may also correlate with several other types of cancers or diseases, where further studies are warranted.
Collapse
|
14
|
Kumar K, Ghosh A. Radiochemistry, Production Processes, Labeling Methods, and ImmunoPET Imaging Pharmaceuticals of Iodine-124. Molecules 2021; 26:E414. [PMID: 33466827 PMCID: PMC7830191 DOI: 10.3390/molecules26020414] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 01/01/2023] Open
Abstract
Target-specific biomolecules, monoclonal antibodies (mAb), proteins, and protein fragments are known to have high specificity and affinity for receptors associated with tumors and other pathological conditions. However, the large biomolecules have relatively intermediate to long circulation half-lives (>day) and tumor localization times. Combining superior target specificity of mAbs and high sensitivity and resolution of the PET (Positron Emission Tomography) imaging technique has created a paradigm-shifting imaging modality, ImmunoPET. In addition to metallic PET radionuclides, 124I is an attractive radionuclide for radiolabeling of mAbs as potential immunoPET imaging pharmaceuticals due to its physical properties (decay characteristics and half-life), easy and routine production by cyclotrons, and well-established methodologies for radioiodination. The objective of this report is to provide a comprehensive review of the physical properties of iodine and iodine radionuclides, production processes of 124I, various 124I-labeling methodologies for large biomolecules, mAbs, and the development of 124I-labeled immunoPET imaging pharmaceuticals for various cancer targets in preclinical and clinical environments. A summary of several production processes, including 123Te(d,n)124I, 124Te(d,2n)124I, 121Sb(α,n)124I, 123Sb(α,3n)124I, 123Sb(3He,2n)124I, natSb(α, xn)124I, natSb(3He,n)124I reactions, a detailed overview of the 124Te(p,n)124I reaction (including target selection, preparation, processing, and recovery of 124I), and a fully automated process that can be scaled up for GMP (Good Manufacturing Practices) production of large quantities of 124I is provided. Direct, using inorganic and organic oxidizing agents and enzyme catalysis, and indirect, using prosthetic groups, 124I-labeling techniques have been discussed. Significant research has been conducted, in more than the last two decades, in the development of 124I-labeled immunoPET imaging pharmaceuticals for target-specific cancer detection. Details of preclinical and clinical evaluations of the potential 124I-labeled immunoPET imaging pharmaceuticals are described here.
Collapse
Affiliation(s)
- Krishan Kumar
- Laboratory for Translational Research in Imaging Pharmaceuticals, The Wright Center of Innovation in Biomedical Imaging, Department of Radiology, The Ohio State University, Columbus, OH 43212, USA;
| | | |
Collapse
|
15
|
Bois F, Noirot C, Dietemann S, Mainta IC, Zilli T, Garibotto V, Walter MA. [ 68Ga]Ga-PSMA-11 in prostate cancer: a comprehensive review. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2020; 10:349-374. [PMID: 33329937 PMCID: PMC7724278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/15/2020] [Indexed: 06/12/2023]
Abstract
Imaging of the prostate-specific membrane antigen (PSMA) has become an important tool for managing patients with recurrent prostate cancer, and one of the most frequently employed radiopharmaceuticals is [68Ga]Ga-PSMA-11. Herein, we summarize the preclinical development and the clinical applications of [68Ga]Ga-PSMA-11 and present side-by-side comparisons with other radiopharmaceuticals or imaging modalities, in order to assist imagers and clinicians in recommending, performing, and interpreting the results of [68Ga]Ga-PSMA-11 PET scans in patients with prostate cancer.
Collapse
Affiliation(s)
- Frédéric Bois
- Division of Nuclear Medicine, Diagnostic Department, University Hospital of GenevaGeneva, Switzerland
| | - Camille Noirot
- Division of Nuclear Medicine, Diagnostic Department, University Hospital of GenevaGeneva, Switzerland
| | - Sébastien Dietemann
- Division of Nuclear Medicine, Diagnostic Department, University Hospital of GenevaGeneva, Switzerland
| | - Ismini C Mainta
- Division of Nuclear Medicine, Diagnostic Department, University Hospital of GenevaGeneva, Switzerland
| | - Thomas Zilli
- Division of Radiation Oncology, Oncology Department, University Hospital of GenevaGeneva, Switzerland
- Faculty of Medicine, University of GenevaGeneva, Switzerland
| | - Valentina Garibotto
- Division of Nuclear Medicine, Diagnostic Department, University Hospital of GenevaGeneva, Switzerland
- Faculty of Medicine, University of GenevaGeneva, Switzerland
| | - Martin A Walter
- Division of Nuclear Medicine, Diagnostic Department, University Hospital of GenevaGeneva, Switzerland
- Faculty of Medicine, University of GenevaGeneva, Switzerland
- Center for Biomedical Imaging (CIBM)Lausanne, Switzerland
| |
Collapse
|
16
|
Liapis V, Tieu W, Rudd SE, Donnelly PS, Wittwer NL, Brown MP, Staudacher AH. Improved non-invasive positron emission tomographic imaging of chemotherapy-induced tumor cell death using Zirconium-89-labeled APOMAB®. EJNMMI Radiopharm Chem 2020; 5:27. [PMID: 33205364 PMCID: PMC7672150 DOI: 10.1186/s41181-020-00109-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 10/19/2020] [Indexed: 01/01/2023] Open
Abstract
Purpose The chimeric monoclonal antibody (mAb) chDAB4 (APOMAB®) targets the Lupus associated (La)/Sjögren Syndrome-B (SSB) antigen, which is over-expressed in tumors but only becomes available for antibody binding in dead tumor cells. Hence, chDAB4 may be used as a novel theranostic tool to distinguish between responders and nonresponders early after chemotherapy. Here, we aimed to ascertain which positron emitter, Zirconium-89 ([89Zr]ZrIV) or Iodine-124 ([124I]I), was best suited to label chDAB4 for post-chemotherapy PET imaging of tumor-bearing mice and to determine which of two different bifunctional chelators provided optimal tumor imaging by PET using [89Zr]ZrIV-labeled chDAB4. Methods C57BL/6 J mice bearing subcutaneous syngeneic tumors of EL4 lymphoma were either untreated or given chemotherapy, then administered radiolabeled chDAB4 after 24 h with its biodistribution examined using PET and organ assay. We compared chDAB4 radiolabeled with [89Zr] ZrIV or [124I] I, or [89Zr]Zr-chDAB4 using either DFO-NCS or DFOSq as a chelator. Results After chemotherapy, [89Zr]Zr-chDAB4 showed higher and prolonged mean (± SD) tumor uptake of 29.5 ± 5.9 compared to 7.8 ± 1.2 for [124I] I -chDAB4. In contrast, antibody uptake in healthy tissues was not affected. Compared to DFO-NCS, DFOSq did not result in significant differences in tumor uptake of [89Zr]Zr-chDAB4 but did alter the tumor:liver ratio in treated mice 3 days after injection in favour of DFOSq (8.0 ± 1.1) compared to DFO-NCS (4.2 ± 0.7). Conclusion ImmunoPET using chDAB4 radiolabeled with residualizing [89Zr] ZrIV rather than [124I] I optimized post-chemotherapy tumor uptake. Further, PET imaging characteristics were improved by DFOSq rather than DFO-NCS. Therefore, the radionuclide/chelator combination of [89Zr] ZrIV and DFOSq is preferred for the imminent clinical evaluation of chDAB4 as a selective tumor cell death radioligand. Supplementary Information Supplementary information accompanies this paper at 10.1186/s41181-020-00109-6.
Collapse
Affiliation(s)
- Vasilios Liapis
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, 5000, Australia.
| | - William Tieu
- School of Medicine, University of Adelaide, Adelaide, SA, 5000, Australia.,Molecular Imaging and Therapy Research Unit (MITRU), South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Stacey E Rudd
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Nicole L Wittwer
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, 5000, Australia
| | - Michael P Brown
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, 5000, Australia.,School of Medicine, University of Adelaide, Adelaide, SA, 5000, Australia.,Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA, 5000, Australia
| | - Alexander H Staudacher
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, 5000, Australia.,School of Medicine, University of Adelaide, Adelaide, SA, 5000, Australia
| |
Collapse
|
17
|
Moskal P, Stępień EŁ. Prospects and Clinical Perspectives of Total-Body PET Imaging Using Plastic Scintillators. PET Clin 2020; 15:439-452. [DOI: 10.1016/j.cpet.2020.06.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
18
|
Yari H, Gali H, Awasthi V. Nanoparticles for Targeting of Prostate Cancer. Curr Pharm Des 2020; 26:5393-5413. [PMID: 32693761 DOI: 10.2174/1381612826666200721001500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/27/2020] [Indexed: 11/22/2022]
Abstract
Prostate cancer (PCa) is the leading cause of death by cancer in men. Because of the drastic decline in the survival rate of PCa patients with advanced/metastatic disease, early diagnosis of disease and therapy without toxic side effects is crucial. Chemotherapy is widely used to control the progression of PCa at the later stages; however, it is associated with off-target toxicities and severe adverse effects due to the lack of specificity. Delivery of therapeutic or diagnostic agents by using targeted nanoparticles is a promising strategy to enhance accuracy and sensitivity of diagnosis of PCa and to increase efficacy and specificity of therapeutic agents. Numerous efforts have been made in past decades to create nanoparticles with different architectural bases for specific delivery payloads to prostate tumors. Major PCa associated cell membrane protein markers identified as targets for such purposes include folate receptor, sigma receptors, transferrin receptor, gastrin-releasing peptide receptor, urokinase plasminogen activator receptor, and prostate specific membrane antigen. Among these markers, prostate specific membrane antigen has emerged as an extremely specific and sensitive targetable marker for designing targeted nanoparticle-based delivery systems for PCa. In this article, we review contemporary advances in design, specificity, and efficacy of nanoparticles functionalized against PCa. Whenever feasible, both diagnostic as well as therapeutic applications are discussed.
Collapse
Affiliation(s)
- Hooman Yari
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Hariprasad Gali
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Vibhudutta Awasthi
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
19
|
Jiménez-Franco LD, Glatting G, Prasad V, Weber WA, Beer AJ, Kletting P. Effect of Tumor Perfusion and Receptor Density on Tumor Control Probability in 177Lu-DOTATATE Therapy: An In Silico Analysis for Standard and Optimized Treatment. J Nucl Med 2020; 62:92-98. [PMID: 32646878 DOI: 10.2967/jnumed.120.245068] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/30/2020] [Indexed: 01/02/2023] Open
Abstract
The aim of this work was to determine a minimal tumor perfusion and receptor density for 177Lu-DOTATATE therapy using physiologically based pharmacokinetic (PBPK) modeling considering, first, a desired tumor control probability (TCP) of 99% and, second, a maximal tolerated biologically effective dose (BEDmax) for organs at risk (OARs) in the treatment of neuroendocrine tumors and meningioma. Methods: A recently developed PBPK model was used. Nine virtual patients (i.e., individualized PBPK models) were used to perform simulations of pharmacokinetics for different combinations of perfusion (0.001-0.1 mL/g/min) and receptor density (1-100 nmol/L). The TCP for each combination was determined for 3 different treatment strategies: a standard treatment (4 cycles of 7.4 GBq and 105 nmol), a treatment maximizing the number of cycles based on BEDmax for red marrow and kidneys, and a treatment having 4 cycles with optimized ligand amount and activity. The red marrow and the kidneys (BEDmax of 2 Gy15 and 40 Gy2.5, respectively) were assumed to be OARs. Additionally, the influence of varying glomerular filtration rates, kidney somatostatin receptor densities, tumor volumes, and release rates was investigated. Results: To achieve a TCP of at least 99% in the standard treatment, a minimal tumor perfusion of 0.036 ± 0.023 mL/g/min and receptor density of 34 ± 20 nmol/L were determined for the 9 virtual patients. With optimization of the number of cycles, the minimum values for perfusion and receptor density were considerably lower, at 0.022 ± 0.012 mL/g/min and 21 ± 11 nmol/L, respectively. However, even better results (perfusion, 0.018 ± 0.009 mL/g/min; receptor density, 18 ± 10 nmol/L) were obtained for strategy 3. The release rate of 177Lu (or labeled metabolites) from tumor cells had the strongest effect on the minimal perfusion and receptor density for standard and optimized treatments. Conclusion: PBPK modeling and simulations represent an elegant approach to individually determine the minimal tumor perfusion and minimal receptor density required to achieve an adequate TCP. This computational method can be used in the radiopharmaceutical development process for ligand and target selection for specific types of tumors. In addition, this method could be used to optimize clinical trials.
Collapse
Affiliation(s)
| | - Gerhard Glatting
- Department of Nuclear Medicine, Ulm University, Ulm, Germany.,Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany; and
| | - Vikas Prasad
- Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Wolfgang A Weber
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Ambros J Beer
- Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Peter Kletting
- Department of Nuclear Medicine, Ulm University, Ulm, Germany .,Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany; and
| |
Collapse
|
20
|
Yoon JK, Park BN, Ryu EK, An YS, Lee SJ. Current Perspectives on 89Zr-PET Imaging. Int J Mol Sci 2020; 21:ijms21124309. [PMID: 32560337 PMCID: PMC7352467 DOI: 10.3390/ijms21124309] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Abstract
89Zr is an emerging radionuclide that plays an essential role in immuno-positron emission tomography (PET) imaging. The long half-life of 89Zr (t1/2 = 3.3 days) is favorable for evaluating the in vivo distribution of monoclonal antibodies. Thus, the use of 89Zr is promising for monitoring antibody-based cancer therapies. Immuno-PET combines the sensitivity of PET with the specificity of antibodies. A number of studies have been conducted to investigate the feasibility of 89Zr immuno-PET imaging for predicting the efficacy of radioimmunotherapy and antibody therapies, imaging target expression, detecting target-expressing tumors, and the monitoring of anti-cancer chemotherapies. In this review, we summarize the current status of PET imaging using 89Zr in both preclinical and clinical studies by highlighting the use of immuno-PET for the targets of high clinical relevance. We also present 89Zr-PET applications other than immuno-PET, such as nanoparticle imaging and cell tracking. Finally, we discuss the limitations and the ongoing research being performed to overcome the remaining hurdles.
Collapse
Affiliation(s)
- Joon-Kee Yoon
- Department of Nuclear Medicine & Molecular Imaging, Ajou University School of Medicine, Worldcup-ro 164, Suwon 16499, Korea; (B.-N.P.); (Y.-S.A.); (S.-J.L.)
- Correspondence: ; Tel.: +82-31-219-4303
| | - Bok-Nam Park
- Department of Nuclear Medicine & Molecular Imaging, Ajou University School of Medicine, Worldcup-ro 164, Suwon 16499, Korea; (B.-N.P.); (Y.-S.A.); (S.-J.L.)
| | - Eun-Kyoung Ryu
- Division of Magnetic Resonance, Korea Basic Science Institute, 162, Yeongudanji-ro, Cheongju 28119, Korea;
| | - Young-Sil An
- Department of Nuclear Medicine & Molecular Imaging, Ajou University School of Medicine, Worldcup-ro 164, Suwon 16499, Korea; (B.-N.P.); (Y.-S.A.); (S.-J.L.)
| | - Su-Jin Lee
- Department of Nuclear Medicine & Molecular Imaging, Ajou University School of Medicine, Worldcup-ro 164, Suwon 16499, Korea; (B.-N.P.); (Y.-S.A.); (S.-J.L.)
| |
Collapse
|
21
|
Abstract
Immuno-positron emission tomography (immunoPET) is a paradigm-shifting molecular imaging modality combining the superior targeting specificity of monoclonal antibody (mAb) and the inherent sensitivity of PET technique. A variety of radionuclides and mAbs have been exploited to develop immunoPET probes, which has been driven by the development and optimization of radiochemistry and conjugation strategies. In addition, tumor-targeting vectors with a short circulation time (e.g., Nanobody) or with an enhanced binding affinity (e.g., bispecific antibody) are being used to design novel immunoPET probes. Accordingly, several immunoPET probes, such as 89Zr-Df-pertuzumab and 89Zr-atezolizumab, have been successfully translated for clinical use. By noninvasively and dynamically revealing the expression of heterogeneous tumor antigens, immunoPET imaging is gradually changing the theranostic landscape of several types of malignancies. ImmunoPET is the method of choice for imaging specific tumor markers, immune cells, immune checkpoints, and inflammatory processes. Furthermore, the integration of immunoPET imaging in antibody drug development is of substantial significance because it provides pivotal information regarding antibody targeting abilities and distribution profiles. Herein, we present the latest immunoPET imaging strategies and their preclinical and clinical applications. We also emphasize current conjugation strategies that can be leveraged to develop next-generation immunoPET probes. Lastly, we discuss practical considerations to tune the development and translation of immunoPET imaging strategies.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
| | - Zachary T Rosenkrans
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Quan-Yong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53705, United States
| |
Collapse
|
22
|
Ermert J, Benešová M, Hugenberg V, Gupta V, Spahn I, Pietzsch HJ, Liolios C, Kopka K. Radiopharmaceutical Sciences. Clin Nucl Med 2020. [DOI: 10.1007/978-3-030-39457-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
23
|
Begum NJ, Glatting G, Wester HJ, Eiber M, Beer AJ, Kletting P. The effect of ligand amount, affinity and internalization on PSMA-targeted imaging and therapy: A simulation study using a PBPK model. Sci Rep 2019; 9:20041. [PMID: 31882829 PMCID: PMC6934468 DOI: 10.1038/s41598-019-56603-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/13/2019] [Indexed: 02/07/2023] Open
Abstract
The aim of this work was to investigate the effect of ligand amount, affinity and internalization of prostate-specific membrane antigen (PSMA)-specific ligands on the activity concentrations for PET/CT imaging and on the absorbed doses for therapy. A physiologically-based pharmacokinetic (PBPK) model for PSMA-specific ligands was implemented. Thirteen virtual patients with metastatic castration-resistant prostate cancer were analysed. Simulations were performed for different combinations of association rates kon (0.1-0.01 L/nmol/min), dissociation rates koff (0.1-0.0001 min-1), internalization rates λint (0.01-0.0001 min-1) and ligand amounts (1-1000 nmol). For imaging the activity was normalized to volume and injected activity (68Ga-PSMA at 1 h). For therapy the absorbed dose was calculated for 7.3 ± 0.3 GBq 177Lu-PSMA. The effect of the investigated parameters on therapy were larger compared to imaging. For imaging, the combination of properties leading to the highest tumour uptake was kon = 0.1 L/nmol/min, koff = 0.01 min-1 for typical ligand amounts (1-10 nmol). For therapy, the higher the internalization rate, the larger was the required ligand amount for optimal tumour-to-kidney ratios. The higher the affinity, the more important was the choice of the optimal ligand amount. PBPK modelling provides insight into the pharmacokinetics of PSMA-specific ligands. Further in silico and in vivo studies are required to verify the influence of the analysed parameters.
Collapse
Affiliation(s)
- Nusrat J Begum
- Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany.
| | - Gerhard Glatting
- Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany.,Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Hans-Jürgen Wester
- Technical University of Munich, Pharmaceutical Radiochemistry, Munich, Germany
| | - Matthias Eiber
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Nuclear Medicine, Munich, Germany
| | - Ambros J Beer
- Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Peter Kletting
- Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany.,Department of Nuclear Medicine, Ulm University, Ulm, Germany
| |
Collapse
|
24
|
Mahzouni P, Shavakhi M. Prostate-Specific Membrane Antigen Expression in Neovasculature of Glioblastoma Multiforme. Adv Biomed Res 2019; 8:18. [PMID: 30993088 PMCID: PMC6425742 DOI: 10.4103/abr.abr_209_18] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: Glioblastoma (GBM) is the most malignant brain tumor with a poor prognosis that can be very difficult to cure, and the current treatment options have no optimal outcomes. Hence, it is essential to find new treatment modalities. Histologically, this tumor has high microvascular density that makes it desirable for vascular target agent drugs. Prostate-specific membrane antigen (PSMA) is a novel antigen with unique features that expresses in the vascular endothelium of some malignant tumors. Materials and Methods: Formalin-fixed paraffin-embedded tissues from sixty patients who underwent GBM tumor resection from 2012 to 2016 were evaluated for the expression of PSMA by immunohistochemistry. Sections were also assessed for the extent and intensity of endothelial staining in tumor microvessels and for clinicopathologic factor correlation. Results: A considerable PSMA expression level was detected in 66% of the cases, and the intensity was strong and moderate in 63%. There was no significant correlation neither between PSMA expression with tumor site, presence of necrosis, and endothelial proliferation nor with age and sex. Conclusion: The expression of PSMA in GBM, as observed in the current study, may suggest a new role of PSMA-targeted therapy and indicate more investigations focused on complementary treatment strategies that specifically target tumor vasculature.
Collapse
Affiliation(s)
- Parvin Mahzouni
- Department of Pathology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mitra Shavakhi
- Department of Pathology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
25
|
Cheal SM, Ruan S, Veach DR, Longo VA, Punzalan BJ, Wu J, Fung EK, Kelly MP, Kirshner JR, Giurleo JT, Ehrlich G, Han AQ, Thurston G, Olson WC, Zanzonico PB, Larson SM, Carrasquillo JA. ImmunoPET Imaging of Endogenous and Transfected Prolactin Receptor Tumor Xenografts. Mol Pharm 2018; 15:2133-2141. [PMID: 29684277 DOI: 10.1021/acs.molpharmaceut.7b01133] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Antibodies labeled with positron-emitting isotopes have been used for tumor detection, predicting which patients may respond to tumor antigen-directed therapy, and assessing pharmacodynamic effects of drug interventions. Prolactin receptor (PRLR) is overexpressed in breast and prostate cancers and is a new target for cancer therapy. We evaluated REGN2878, an anti-PRLR monoclonal antibody, as an immunoPET reagent. REGN2878 was labeled with Zr-89 after conjugation with desferrioxamine B or labeled with I-131/I-124. In vitro determination of the half-maximal inhibitory concentration (IC50) of parental REGN2878, DFO-REGN2878, and iodinated REGN2878 was performed by examining the effect of the increasing amounts of these on uptake of trace-labeled I-131 REGN2878. REGN1932, a non-PRLR binding antibody, was used as a control. Imaging and biodistribution studies were performed in mice bearing tumor xenografts with various expression levels of PRLR, including MCF-7, transfected MCF-7/PRLR, PC3, and transfected PC3/PRLR and T4D7v11 cell lines. The specificity of uptake in tumors was evaluated by comparing Zr-89 REGN2878 and REGN1932, and in vivo competition compared Zr-89 REGN2878 uptake in tumor xenografts with and without prior injection of 2 mg of nonradioactive REGN2878. The competition binding assay of DFO-REGN2878 at ratios of 3.53-5.77 DFO per antibody showed IC50 values of 0.4917 and 0.7136 nM, respectively, compared to 0.3455 nM for parental REGN2878 and 0.3343 nM for I-124 REGN2878. Imaging and biodistribution studies showed excellent targeting of Zr-89 REGN2878 in PRLR-positive xenografts at delayed times of 189 h (presented as mean ± 1 SD, percent injected activity per mL (%IA/mL) 74.6 ± 33.8%IA/mL). In contrast, MCF-7/PRLR tumor xenografts showed a low uptake (7.0 ± 2.3%IA/mL) of control Zr-89 REGN1932 and a very low uptake and rapid clearance of I-124 REGN2878 (1.4 ± 0.6%IA/mL). Zr-89 REGN2878 has excellent antigen-specific targeting in various PRLR tumor xenograft models. We estimated, using image-based kinetic modeling, that PRLR antigen has a very rapid in vivo turnover half-life of ∼14 min from the cell membrane. Despite relatively modest estimated tumor PRLR expression numbers, PRLR-expressing cells have shown final retention of the Zr-89 REGN2878 antibody, with an uptake that appeared to be related to PRLR expression. This reagent has the potential to be used in clinical trials targeting PRLR.
Collapse
Affiliation(s)
- Sarah M Cheal
- Department of Radiology , Memorial Sloan Kettering Cancer Center (MSK) , New York , NY 10065 , United States.,Molecular Pharmacology Program , MSK , New York , NY 10065 , United States.,Center for Targeted Radioimmunotherapy and Diagnosis , Ludwig Center for Cancer Immunotherapy , New York , NY 10065 , United States
| | - Shutian Ruan
- Department of Radiology , Memorial Sloan Kettering Cancer Center (MSK) , New York , NY 10065 , United States.,Molecular Pharmacology Program , MSK , New York , NY 10065 , United States.,Center for Targeted Radioimmunotherapy and Diagnosis , Ludwig Center for Cancer Immunotherapy , New York , NY 10065 , United States
| | - Darren R Veach
- Department of Radiology , Memorial Sloan Kettering Cancer Center (MSK) , New York , NY 10065 , United States.,Molecular Pharmacology Program , MSK , New York , NY 10065 , United States
| | - Valerie A Longo
- Small-Animal Imaging Core Facility , MSK , New York , NY 10065 , United States
| | - Blesida J Punzalan
- Department of Radiology , Memorial Sloan Kettering Cancer Center (MSK) , New York , NY 10065 , United States.,Molecular Pharmacology Program , MSK , New York , NY 10065 , United States
| | - Jiong Wu
- Department of Radiology , Memorial Sloan Kettering Cancer Center (MSK) , New York , NY 10065 , United States
| | - Edward K Fung
- Department of Radiology , Memorial Sloan Kettering Cancer Center (MSK) , New York , NY 10065 , United States.,Department of Medical Physics , MSK , New York , NY 10065 , United States
| | - Marcus P Kelly
- Regeneron Pharmaceuticals, Inc. , Tarrytown , NY 10591 , United States
| | | | - Jason T Giurleo
- Regeneron Pharmaceuticals, Inc. , Tarrytown , NY 10591 , United States
| | - George Ehrlich
- Regeneron Pharmaceuticals, Inc. , Tarrytown , NY 10591 , United States
| | - Amy Q Han
- Regeneron Pharmaceuticals, Inc. , Tarrytown , NY 10591 , United States
| | - Gavin Thurston
- Regeneron Pharmaceuticals, Inc. , Tarrytown , NY 10591 , United States
| | - William C Olson
- Regeneron Pharmaceuticals, Inc. , Tarrytown , NY 10591 , United States
| | - Pat B Zanzonico
- Molecular Pharmacology Program , MSK , New York , NY 10065 , United States.,Small-Animal Imaging Core Facility , MSK , New York , NY 10065 , United States.,Department of Medical Physics , MSK , New York , NY 10065 , United States
| | - Steven M Larson
- Department of Radiology , Memorial Sloan Kettering Cancer Center (MSK) , New York , NY 10065 , United States.,Molecular Pharmacology Program , MSK , New York , NY 10065 , United States.,Center for Targeted Radioimmunotherapy and Diagnosis , Ludwig Center for Cancer Immunotherapy , New York , NY 10065 , United States.,Department of Radiology , Weill Cornell Medical Center , New York , NY 10065 , United States
| | - Jorge A Carrasquillo
- Department of Radiology , Memorial Sloan Kettering Cancer Center (MSK) , New York , NY 10065 , United States.,Molecular Pharmacology Program , MSK , New York , NY 10065 , United States.,Center for Targeted Radioimmunotherapy and Diagnosis , Ludwig Center for Cancer Immunotherapy , New York , NY 10065 , United States.,Department of Radiology , Weill Cornell Medical Center , New York , NY 10065 , United States
| |
Collapse
|
26
|
Burvenich IJG, Parakh S, Parslow AC, Lee ST, Gan HK, Scott AM. Receptor Occupancy Imaging Studies in Oncology Drug Development. AAPS JOURNAL 2018. [DOI: 10.1208/s12248-018-0203-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
27
|
Lenzo NP, Meyrick D, Turner JH. Review of Gallium-68 PSMA PET/CT Imaging in the Management of Prostate Cancer. Diagnostics (Basel) 2018; 8:E16. [PMID: 29439481 PMCID: PMC5871999 DOI: 10.3390/diagnostics8010016] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/01/2018] [Accepted: 02/01/2018] [Indexed: 01/13/2023] Open
Abstract
Over 90% of prostate cancers over-express prostate specific membrane antigen (PSMA) and these tumor cells may be accurately targeted for diagnosis by 68Ga-PSMA-positron emission tomography/computed tomography (68Ga-PSMA-PET/CT) imaging. This novel molecular imaging modality appears clinically to have superseded CT, and appears superior to MR imaging, for the detection of metastatic disease. 68Ga-PSMA PET/CT has the ability to reliably stage prostate cancer at presentation and can help inform an optimal treatment approach. Novel diagnostic applications of 68Ga-PSMA PET/CT include guiding biopsy to improve sampling accuracy, and guiding surgery and radiotherapy. In addition to facilitating the management of metastatic castrate resistant prostate cancer (mCRPC), 68Ga-PSMA can select patients who may benefit from targeted systemic radionuclide therapy. 68Ga-PSMA is the diagnostic positron-emitting theranostic pair with the beta emitter Lutetium-177 PSMA (177Lu-PSMA) and alpha-emitter Actinium-225 PSMA (225Ac-PSMA) which can both be used to treat PSMA-avid metastases of prostate cancer in the molecular tumor-targeted approach of theranostic nuclear oncology.
Collapse
Affiliation(s)
- Nat P Lenzo
- Nuclear Oncology, Theranostics Australia, 106/1 Silas Street, Richmond Quarter Building, East Fremantle, WA 6158, Australia.
- School of Medicine and Pharmacology, University of Western Australia, 35 Stirling Highway, Nedlands, WA 6009, Australia.
| | - Danielle Meyrick
- Nuclear Oncology, Theranostics Australia, 106/1 Silas Street, Richmond Quarter Building, East Fremantle, WA 6158, Australia.
| | - J Harvey Turner
- School of Medicine and Pharmacology, University of Western Australia, 35 Stirling Highway, Nedlands, WA 6009, Australia.
| |
Collapse
|
28
|
Osborne JR, Kalidindi TM, Punzalan BJ, Gangangari K, Spratt DE, Weber WA, Larson SM, Pillarsetty NVK. Repeatability of [ 68Ga]DKFZ11-PSMA PET Scans for Detecting Prostate-specific Membrane Antigen-positive Prostate Cancer. Mol Imaging Biol 2017; 19:944-951. [PMID: 28534214 PMCID: PMC5664162 DOI: 10.1007/s11307-017-1091-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE We studied the effect of varying specific activity of [68Ga]DKFZ-PSMA11 ([68Ga]DP11) on repeated imaging of prostate-specific membrane antigen-positive (PSMA+) xenograft tumors. PROCEDURES Athymic nude mice bearing PC3-PIP (PSMA+) and PC3 (PSMA-) bilateral flank tumors were assessed to study intra- and inter-day repeatability of [68Ga]DP11 imaging in mice administered [68Ga]DP11 or [67Ga]DP11 (as a dilution tracer) using imaging and biodistribution studies. RESULTS Region of interest (ROI) analysis of the [68Ga]DP11 imaging study indicated that the uptake was constant on the same day or consecutive days. Prior imaging with [68Ga]DP11 did not significantly influence the subsequent uptake of [68Ga]DP11. Uptake of [68Ga]DP11 (60 min) and [67Ga]DP11 (24 h) in PC3-PIP tumors was 12.37 ± 4.19 %ID/g and 12.49 ± 6.88 %ID/g, respectively; [68Ga]DP11 was 13.83 ± 3.77 and 17.76 ± 1.84 on same-day and 15.98 ± 5.82 %ID/g on second-day imaging. CONCLUSIONS This study demonstrates that [68Ga]DP11, in a given PSMA+ lesion, is constant under several same-day or serial-day imaging conditions.
Collapse
Affiliation(s)
- Joseph R. Osborne
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Radiology, Weill Cornell Medical College, New York, NY
| | - Teja M. Kalidindi
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Blesida J. Punzalan
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kishore Gangangari
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Chemistry, Hunter College and PhD Program in Chemistry, The Graduate Center of the City University of New York, New York, NY
| | - Daniel E. Spratt
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
| | - Wolfgang A. Weber
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Steven M. Larson
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Radiology, Weill Cornell Medical College, New York, NY
| | - Naga Vara Kishore Pillarsetty
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Radiology, Weill Cornell Medical College, New York, NY
| |
Collapse
|
29
|
Molecular imaging in drug development: Update and challenges for radiolabeled antibodies and nanotechnology. Methods 2017; 130:23-35. [DOI: 10.1016/j.ymeth.2017.07.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/08/2017] [Accepted: 07/18/2017] [Indexed: 01/01/2023] Open
|