1
|
Badier L, Quelven I. Zirconium 89 and Copper 64 for ImmunoPET: From Antibody Bioconjugation and Radiolabeling to Molecular Imaging. Pharmaceutics 2024; 16:882. [PMID: 39065579 PMCID: PMC11279968 DOI: 10.3390/pharmaceutics16070882] [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: 04/30/2024] [Revised: 06/15/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Immunotherapy has transformed cancer treatment. Nevertheless, given the heterogeneity of clinical efficacy, the multiplicity of treatment options available and the possibility of serious adverse effects, selecting the most effective treatment has become the greatest challenge. Molecular imaging offers an attractive way for this purpose. ImmunoPET provides specific imaging with positron emission tomography (PET) using monoclonal antibodies (mAb) or its fragments as vector. By combining the high targeting specificity of mAb and the sensitivity of PET technique, immunoPET could noninvasively and dynamically reveal tumor antigens expression and provide theranostic tools of several types of malignancies. Because of their slow kinetics, mAbs require radioelements defined by a consistent half-life. Zirconium 89 (89Zr) and Copper 64 (64Cu) are radiometals with half-lives suitable for mAb labeling. Radiolabeling with a radiometal requires the prior use of a bifunctional chelate agent (BFCA) to functionalize mAb for radiometal chelation, in a second step. There are a number of BFCA available and much research is focused on antibody functionalization techniques or on developing the optimum chelating agent depending the selected radiometal. In this manuscript, we present a critical account of radiochemical techniques with radionuclides 89Zr and 64Cu and their applications in preclinical and clinical immuno-PET imaging.
Collapse
Affiliation(s)
| | - Isabelle Quelven
- Toulouse NeuroImaging Center (ToNIC), INSERM/UPS UMR 1214, University Hospital of Toulouse-Purpan, CEDEX 3, 31024 Toulouse, France;
| |
Collapse
|
2
|
He H, Qi X, Fu H, Xu J, Zheng Q, Chen L, Zhang Y, Hua H, Xu W, Xu Z, Chen X, You Q, Lin J, Huang G, Mao Y, Yu C. Imaging diagnosis and efficacy monitoring by [ 89Zr]Zr-DFO-KN035 immunoPET in patients with PD-L1-positive solid malignancies. Theranostics 2024; 14:392-405. [PMID: 38164149 PMCID: PMC10750192 DOI: 10.7150/thno.87243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 11/08/2023] [Indexed: 01/03/2024] Open
Abstract
Rationale: Although programmed death-ligand 1 (PD-L1) inhibitors have achieved efficacy in cancer therapy, their response rate is low. Differences in the prognosis of patients with cancer under anti-PD-L1 treatment are related to the PD-L1 level in tumors. Accurate PD-L1 detection can optimize the accuracy of tumor immunotherapy and avoid ineffective clinical diagnosis and treatments. Methods: We investigated the imaging efficiency and therapy monitoring capacity of [89Zr]Zr-DFO-KN035 immunoPET for tumors. We labeled the monodomain anti-PD-L1 antibody KN035 with the radionuclide zirconium-89 and used this tracer for PET imaging. [89Zr]Zr-DFO-KN035 uptakes in patients with PD-L1-positive tumors, including primary and metastatic tumors, as well as in normal tissues, were comparatively assessed by using positron emission tomography/computed tomography imaging. Results: In PD-L1-positive patients, [89Zr]Zr-DFO-KN035 was sensitive in tumor-targeting imaging and could detect multiple metastatic foci, including multiple bone metastases (tumor-to-muscle ratios of 7.102 and 6.118 at 55 and 120 h, respectively) and lymph-node metastases (tumor-to-muscle ratios of 11.346 and 6.542 at 55 and 120 h, respectively). The needed radioactive dose of [89Zr]Zr-DFO-KN035 (55.5-92.5 MBq) used in this study was considerably lower than that of [18F]FDG (370-555 MBq). [89Zr]Zr-DFO-KN035 monitored and predicted the site of adverse reactions in antitumor immunotherapy. Moreover, after antitumor treatment, [89Zr]Zr-DFO-KN035 enabled observational imaging for therapeutic efficacy evaluation, which can help predict patient prognosis. Conclusion: [89Zr]Zr-DFO-KN035 can be used for the diagnosis and therapy monitoring of PD-L1-positive tumors and provide noninvasive and comprehensive observations for tumor diagnostic imaging, prognosis prediction, and efficacy evaluation.
Collapse
Affiliation(s)
- Huihui He
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University; Wuxi, China
| | - Xiaowei Qi
- Department of Pathology, Affiliated Hospital of Jiangnan University; Wuxi, China
| | - Haitian Fu
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University; Wuxi, China
| | - Jianfeng Xu
- Dongcheng AMS Pharmaceutical Co., Ltd.; Nanjing, China
| | - Qihuang Zheng
- Center of Radiological Imaging, College of Medicine, Indiana University, Indiana, USA
| | - Liping Chen
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University; Wuxi, China
| | - Yu Zhang
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University; Wuxi, China
| | - Haiying Hua
- Department of Oncology, Affiliated Hospital of Jiangnan University; Wuxi, China
| | - Wenhuan Xu
- Department of Oncology, Affiliated Hospital of Jiangnan University; Wuxi, China
| | - Zhenyu Xu
- Department of Oncology, Affiliated Hospital of Jiangnan University; Wuxi, China
| | - Xiaoping Chen
- Department of Oncology, Affiliated Hospital of Jiangnan University; Wuxi, China
| | - Qingjun You
- Institute of Oncology, Affiliated Hospital of Jiangnan University; Wuxi, China
| | - Jianguo Lin
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine; Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences; Shanghai, China
| | - Yong Mao
- Department of Oncology, Affiliated Hospital of Jiangnan University; Wuxi, China
| | - Chunjing Yu
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University; Wuxi, China
| |
Collapse
|
3
|
Arroyo A, Lyashchenko SK, Lewis JS. Methods for the Production of Radiolabeled Bioagents for ImmunoPET. Methods Mol Biol 2024; 2729:117-142. [PMID: 38006494 PMCID: PMC11330323 DOI: 10.1007/978-1-0716-3499-8_8] [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: 11/27/2023]
Abstract
Immunoglobulin-based positron emission tomography (ImmunoPET) is making increasingly significant contributions to the nuclear imaging toolbox. The exquisite specificity of antibodies combined with the high-resolution imaging of PET enables clinicians and researchers to localize diseases, especially cancer, with a high degree of spatial certainty. This review focuses on the radiopharmaceutical preparation necessary to obtain those images-the work behind the scenes, which occurs even before the patient or animal is injected with the radioimmunoconjugate. The focus of this methods review will be the chelation of four radioisotopes to their most common and clinically relevant chelators.
Collapse
Affiliation(s)
- Alejandro Arroyo
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Serge K Lyashchenko
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Radiochemistry and Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Radiochemistry and Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
4
|
Aguilar-Calvo P, Malik A, Sandoval DR, Barback C, Orrù CD, Standke HG, Thomas OR, Dwyer CA, Pizzo DP, Bapat J, Soldau K, Ogawa R, Riley MB, Nilsson KPR, Kraus A, Caughey B, Iliff JJ, Vera DR, Esko JD, Sigurdson CJ. Neuronal Ndst1 depletion accelerates prion protein clearance and slows neurodegeneration in prion infection. PLoS Pathog 2023; 19:e1011487. [PMID: 37747931 PMCID: PMC10586673 DOI: 10.1371/journal.ppat.1011487] [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: 06/16/2023] [Revised: 10/19/2023] [Accepted: 08/11/2023] [Indexed: 09/27/2023] Open
Abstract
Select prion diseases are characterized by widespread cerebral plaque-like deposits of amyloid fibrils enriched in heparan sulfate (HS), a abundant extracellular matrix component. HS facilitates fibril formation in vitro, yet how HS impacts fibrillar plaque growth within the brain is unclear. Here we found that prion-bound HS chains are highly sulfated, and that the sulfation is essential for accelerating prion conversion in vitro. Using conditional knockout mice to deplete the HS sulfation enzyme, Ndst1 (N-deacetylase / N-sulfotransferase) from neurons or astrocytes, we investigated how reducing HS sulfation impacts survival and prion aggregate distribution during a prion infection. Neuronal Ndst1-depleted mice survived longer and showed fewer and smaller parenchymal plaques, shorter fibrils, and increased vascular amyloid, consistent with enhanced aggregate transit toward perivascular drainage channels. The prolonged survival was strain-dependent, affecting mice infected with extracellular, plaque-forming, but not membrane bound, prions. Live PET imaging revealed rapid clearance of recombinant prion protein monomers into the CSF of neuronal Ndst1- deficient mice, neuronal, further suggesting that HS sulfate groups hinder transit of extracellular prion protein monomers. Our results directly show how a host cofactor slows the spread of prion protein through the extracellular space and identify an enzyme to target to facilitate aggregate clearance.
Collapse
Affiliation(s)
| | - Adela Malik
- Department of Pathology, UC San Diego, La Jolla, California, United States of America
| | - Daniel R. Sandoval
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, California, United States of America
| | - Christopher Barback
- Department of Radiology, UC San Diego, La Jolla, California, United States of America
| | - Christina D. Orrù
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Heidi G. Standke
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Olivia R. Thomas
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Chrissa A. Dwyer
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, California, United States of America
| | - Donald P. Pizzo
- Department of Pathology, UC San Diego, La Jolla, California, United States of America
| | - Jaidev Bapat
- Department of Pathology, UC San Diego, La Jolla, California, United States of America
| | - Katrin Soldau
- Department of Pathology, UC San Diego, La Jolla, California, United States of America
| | - Ryotaro Ogawa
- Department of Radiology, UC San Diego, La Jolla, California, United States of America
| | - Mckenzie B. Riley
- Department of Neurology, University of Alabama, Birmingham, Alabama, United States of America
| | - K. Peter R. Nilsson
- Department of Physics, Chemistry, and Biology, Linköping University, Linköping, Sweden
| | - Allison Kraus
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Byron Caughey
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Jeffrey J. Iliff
- VISN 20 NW Mental Illness Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, United States of America
- Department of Psychiatry and Behavioral Science, Department of Neurology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - David R. Vera
- Department of Radiology, UC San Diego, La Jolla, California, United States of America
| | - Jeffrey D. Esko
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, California, United States of America
| | - Christina J. Sigurdson
- Department of Pathology, UC San Diego, La Jolla, California, United States of America
- Department of Medicine, UC San Diego, La Jolla, California, United States of America
- Department of Pathology, Microbiology, and Immunology, UC Davis, Davis, California, United States of America
| |
Collapse
|
5
|
Ishibashi M, Takahashi M, Yamaya T, Imai Y. Current and Future PET Imaging for Multiple Myeloma. Life (Basel) 2023; 13:1701. [PMID: 37629558 PMCID: PMC10455506 DOI: 10.3390/life13081701] [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: 06/07/2023] [Revised: 07/26/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Positron emission tomography (PET) is an imaging modality used for the noninvasive assessment of tumor staging and response to therapy. PET with 18F labeled fluorodeoxyglucose (18F-FDG PET) is widely used to assess the active and inactive lesions in patients with multiple myeloma (MM). Despite the availability of 18F-FDG PET for the management of MM, PET imaging is less sensitive than next-generation flow cytometry and sequencing. Therefore, the novel PET radiotracers 64Cu-LLP2A, 68Ga-pentixafor, and 89Zr-daratumumab have been developed to target the cell surface antigens of MM cells. Furthermore, recent studies attempted to visualize the tumor-infiltrating lymphocytes using PET imaging in patients with cancer to investigate their prognostic effect; however, these studies have not yet been performed in MM patients. This review summarizes the recent studies on PET with 18F-FDG and novel radiotracers for the detection of MM and the resulting preclinical research using MM mouse models and clinical studies. Novel PET technologies may be useful for developing therapeutic strategies for MM in the future.
Collapse
Affiliation(s)
- Mariko Ishibashi
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo 113-8602, Japan;
| | - Miwako Takahashi
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (M.T.); (T.Y.)
| | - Taiga Yamaya
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (M.T.); (T.Y.)
| | - Yoichi Imai
- Department of Hematology and Oncology, Dokkyo Medical University, Tochigi 321-0293, Japan
| |
Collapse
|
6
|
Métivier C, Le Saëc P, Gaschet J, Chauvet C, Marionneau-Lambot S, Hofgaard PO, Bogen B, Pineau J, Le Bris N, Tripier R, Alliot C, Haddad F, Chérel M, Chouin N, Faivre-Chauvet A, Rbah-Vidal L. Preclinical Evaluation of a 64Cu-Based Theranostic Approach in a Murine Model of Multiple Myeloma. Pharmaceutics 2023; 15:1817. [PMID: 37514004 PMCID: PMC10385603 DOI: 10.3390/pharmaceutics15071817] [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: 05/05/2023] [Revised: 06/16/2023] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
Although the concept of theranostics is neither new nor exclusive to nuclear medicine, it is a particularly promising approach for the future of nuclear oncology. This approach is based on the use of molecules targeting specific biomarkers in the tumour or its microenvironment, associated with optimal radionuclides which, depending on their emission properties, allow the combination of diagnosis by molecular imaging and targeted radionuclide therapy (TRT). Copper-64 has suitable decay properties (both β+ and β- decays) for PET imaging and potentially for TRT, making it both an imaging and therapy agent. We developed and evaluated a theranostic approach using a copper-64 radiolabelled anti-CD138 antibody, [64Cu]Cu-TE1PA-9E7.4 in a MOPC315.BM mouse model of multiple myeloma. PET imaging using [64Cu]Cu-TE1PA-9E7.4 allows for high-resolution PET images. Dosimetric estimation from ex vivo biodistribution data revealed acceptable delivered doses to healthy organs and tissues, and a very encouraging tumour absorbed dose for TRT applications. Therapeutic efficacy resulting in delayed tumour growth and increased survival without inducing major or irreversible toxicity has been observed with 2 doses of 35 MBq administered at a 2-week interval. Repeated injections of [64Cu]Cu-TE1PA-9E7.4 are safe and can be effective for TRT application in this syngeneic preclinical model of MM.
Collapse
Affiliation(s)
- Cassandra Métivier
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44007 Nantes, France
| | - Patricia Le Saëc
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44007 Nantes, France
| | - Joëlle Gaschet
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44007 Nantes, France
| | - Catherine Chauvet
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44007 Nantes, France
| | | | - Peter O Hofgaard
- Department of Immunology, Oslo University Hospital, 04024 Oslo, Norway
| | - Bjarne Bogen
- Department of Immunology, Oslo University Hospital, 04024 Oslo, Norway
| | - Julie Pineau
- Univ. Brest, UMR CNRS 6521 CEMCA, 6 Avenue Victor le Gorgeu, 29200 Brest, France
| | - Nathalie Le Bris
- Univ. Brest, UMR CNRS 6521 CEMCA, 6 Avenue Victor le Gorgeu, 29200 Brest, France
| | - Raphaël Tripier
- Univ. Brest, UMR CNRS 6521 CEMCA, 6 Avenue Victor le Gorgeu, 29200 Brest, France
| | - Cyrille Alliot
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44007 Nantes, France
- GIP ARRONAX, 44800 Saint-Herblain, France
| | - Férid Haddad
- IMT Atlantique, Nantes Université, Subatech, 44307 Nantes, France
- GIP ARRONAX, 44800 Saint-Herblain, France
| | - Michel Chérel
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44007 Nantes, France
- Nuclear Medicine Department, ICO-René Gauducheau Cancer Center, 44800 Saint-Herblain, France
| | - Nicolas Chouin
- Nantes Université, Inserm, CNRS, Université d'Angers, Oniris, CRCI2NA, 44007 Nantes, France
| | - Alain Faivre-Chauvet
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, CHU Nantes, 44007 Nantes, France
| | - Latifa Rbah-Vidal
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44007 Nantes, France
| |
Collapse
|
7
|
Benfante V, Stefano A, Ali M, Laudicella R, Arancio W, Cucchiara A, Caruso F, Cammarata FP, Coronnello C, Russo G, Miele M, Vieni A, Tuttolomondo A, Yezzi A, Comelli A. An Overview of In Vitro Assays of 64Cu-, 68Ga-, 125I-, and 99mTc-Labelled Radiopharmaceuticals Using Radiometric Counters in the Era of Radiotheranostics. Diagnostics (Basel) 2023; 13:diagnostics13071210. [PMID: 37046428 PMCID: PMC10093267 DOI: 10.3390/diagnostics13071210] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/11/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023] Open
Abstract
Radionuclides are unstable isotopes that mainly emit alpha (α), beta (β) or gamma (γ) radiation through radiation decay. Therefore, they are used in the biomedical field to label biomolecules or drugs for diagnostic imaging applications, such as positron emission tomography (PET) and/or single-photon emission computed tomography (SPECT). A growing field of research is the development of new radiopharmaceuticals for use in cancer treatments. Preclinical studies are the gold standard for translational research. Specifically, in vitro radiopharmaceutical studies are based on the use of radiopharmaceuticals directly on cells. To date, radiometric β- and γ-counters are the only tools able to assess a preclinical in vitro assay with the aim of estimating uptake, retention, and release parameters, including time- and dose-dependent cytotoxicity and kinetic parameters. This review has been designed for researchers, such as biologists and biotechnologists, who would like to approach the radiobiology field and conduct in vitro assays for cellular radioactivity evaluations using radiometric counters. To demonstrate the importance of in vitro radiopharmaceutical assays using radiometric counters with a view to radiogenomics, many studies based on 64Cu-, 68Ga-, 125I-, and 99mTc-labeled radiopharmaceuticals have been revised and summarized in this manuscript.
Collapse
Affiliation(s)
- Viviana Benfante
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 90015 Cefalù, Italy
| | - Alessandro Stefano
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 90015 Cefalù, Italy
| | - Muhammad Ali
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy
| | | | - Walter Arancio
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
| | - Antonino Cucchiara
- Department of Diagnostic and Therapeutic Services, IRCCS-ISMETT (Mediterranean Institute for Transplantation and Advanced Specialized Therapies), Via Tricomi 5, 90127 Palermo, Italy
| | - Fabio Caruso
- Department of Diagnostic and Therapeutic Services, IRCCS-ISMETT (Mediterranean Institute for Transplantation and Advanced Specialized Therapies), Via Tricomi 5, 90127 Palermo, Italy
| | - Francesco Paolo Cammarata
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 90015 Cefalù, Italy
| | - Claudia Coronnello
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Giorgio Russo
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 90015 Cefalù, Italy
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Monica Miele
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
| | - Alessandra Vieni
- Department of Diagnostic and Therapeutic Services, IRCCS-ISMETT (Mediterranean Institute for Transplantation and Advanced Specialized Therapies), Via Tricomi 5, 90127 Palermo, Italy
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy
| | - Anthony Yezzi
- Department of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Albert Comelli
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| |
Collapse
|
8
|
Sinomenine ameliorates adjuvant-induced arthritis by inhibiting the autophagy/NETosis/inflammation axis. Sci Rep 2023; 13:3933. [PMID: 36894604 PMCID: PMC9998614 DOI: 10.1038/s41598-023-30922-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Studies have found that neutrophil extracellular traps (NETs) which are the specific dying form of neutrophil upon activation have fundamental role in the rheumatoid arthritis onset and progression. The purpose of this study was to explore the therapeutic effect of Sinomenine on adjuvant-induced arthritis in mice, and the neutrophil activities regulated by Sinomenine. The rheumatoid arthritis model was established by local injection of adjuvant and the Sinomenine treatment was administered orally for 30 days, during which, arthritic scores were evaluated and the joint diameter was measured to determine disease progression. The joint tissues and serum were acquired for further tests after sacrifice. Cytometric beads assay was performed to measure the concentration of cytokines. For paraffin-embedded ankle tissues, hematoxylin and erosin staining and Safranin O-fast staining were adopted to monitor the tissue changes of joint. In order to analyze the inflammation, NETs and autophagy of neutrophils in vivo, immunohistochemistry assays were applied to detect the protein expression levels in the local joints. To describe the effect brought by Sinomenine on inflammation, autophagy and NETs in vitro, the western blotting and the immunofluorescence assays were performed. The joint symptoms of the adjuvant induced arthritis were alleviated by the Sinomenine treatment significantly in terms of the ankle diameter and scores. The improvement of local histopathology changes and decrease of inflammatory cytokines in the serum also confirmed the efficacy. The expression levels of interleukin-6, P65 and p-P65 in the ankle areas of mice were remarkably reduced by Sinomenine. Compared with the model group, the decreased expression levels of lymphocyte antigen 6 complex and myeloperoxidase in the Sinomenine treating group showed the inhibitory effect of Sinomenine on the neutrophil migration. The expression of protein arginine deiminase type 4 (PAD4), ctrullinated histone H3 (CitH3) and microtubule-associated protein 1 light chain 3B (LC3B) had the similar tendency. Upon activation of lipopolysaccharide (LPS) in vitro, Sinomenine suppressed the phosphorylation of P65, extracellular signal-regulated kinase (ERK) and P38 of neutrophil. Meanwhile, Sinomenine inhibited NETs formation induced by phorbol 12-myristate 13-acetate (PMA), which were demonstrated by the decreased expression of neutrophil elastase (NE), PAD4 and CitH3. Sinomenine also inhibited PMA-induced autophagy in vitro based on the changes of Beclin-1 and LC3B. Sinomenine has good efficacy in treating adjuvant induced arthritis via regulating neutrophil activities. Apart from inhibiting activation of nuclear factor kappa-B (NF-κB) and mitogen-activated protein kinase (MAPK) pathways, the mechanism includes suppression of NETs formation via autophagy inhibition.
Collapse
|
9
|
Damerow H, Cheng X, von Kiedrowski V, Schirrmacher R, Wängler B, Fricker G, Wängler C. Toward Optimized 89Zr-Immuno-PET: Side-by-Side Comparison of [ 89Zr]Zr-DFO-, [ 89Zr]Zr-3,4,3-(LI-1,2-HOPO)- and [ 89Zr]Zr-DFO*-Cetuximab for Tumor Imaging: Which Chelator Is the Most Suitable? Pharmaceutics 2022; 14:pharmaceutics14102114. [PMID: 36297549 PMCID: PMC9611803 DOI: 10.3390/pharmaceutics14102114] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/21/2022] Open
Abstract
89Zr represents a highly favorable positron emitter for application in immuno-PET (Positron Emission Tomography) imaging. Clinically, the 89Zr4+ ion is introduced into antibodies by complexation with desferrioxamine B. However, producing complexes of limited kinetic inertness. Therefore, several new chelators for 89Zr introduction have been developed over the last years. Of these, the direct comparison of the most relevant ones for clinical translation, DFO* and 3,4,3-(LI-1,2-HOPO), is still missing. Thus, we directly compared DFO with DFO* and 3,4,3-(LI-1,2-HOPO) immunoconjugates to identify the most suitable agent stable 89Zr-complexation. The chelators were introduced into cetuximab, and an optical analysis method was developed, enabling the efficient quantification of derivatization sites per protein. The cetuximab conjugates were efficiently obtained and radiolabeled with 89Zr at 37 °C within 30 min, giving the [89Zr]Zr-cetuximab derivatives in high radiochemical yields and purities of >99% as well as specific activities of 50 MBq/mg. The immunoreactive fraction of all 89Zr-labeled cetuximab derivatives was determined to be in the range of 86.5−88.1%. In vivo PET imaging and ex vivo biodistribution studies in tumor-bearing animals revealed a comparable and significantly higher kinetic inertness for both [89Zr]Zr-3,4,3-(LI-1,2-HOPO)-cetuximab and [89Zr]Zr-DFO*-cetuximab, compared to [89Zr]Zr-DFO-cetuximab. Of these, [89Zr]Zr-DFO*-cetuximab showed a considerably more favorable pharmacokinetic profile with significantly lower liver and spleen retention than [89Zr]Zr-3,4,3-(LI-1,2-HOPO)-cetuximab. Since [89Zr]Zr-DFO* demonstrates a very high kinetic inertness, paired with a highly favorable pharmacokinetic profile of the resulting antibody conjugate, DFO* currently represents the most suitable chelator candidate for stable 89Zr-radiolabeling of antibodies and clinical translation.
Collapse
Affiliation(s)
- Helen Damerow
- Biomedical Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Xia Cheng
- Molecular Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Valeska von Kiedrowski
- Molecular Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Ralf Schirrmacher
- Division of Oncologic Imaging, Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Björn Wängler
- Molecular Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Carmen Wängler
- Biomedical Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Correspondence:
| |
Collapse
|
10
|
Zhang S, Shang J, Ye W, Zhao T, Xu H, Zeng H, Wang L. Recent developments on the application of molecular probes in multiple myeloma: Beyond [18F]FDG. Front Bioeng Biotechnol 2022; 10:920882. [PMID: 36091426 PMCID: PMC9459033 DOI: 10.3389/fbioe.2022.920882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple myeloma (MM) is a neoplastic plasma cell proliferative disorder characterized by various osteolytic bone destruction as a radiological morphological marker. Functional imaging, particularly nuclear medicine imaging, is a promising method to visualize disease processes before the appearance of structural changes by targeting specific biomarkers related to metabolism ability, tumor microenvironment as well as neoplastic receptors. In addition, by targeting particular antigens with therapeutic antibodies, immuno-PET imaging can support the development of personalized theranostics. At present, various imaging agents have been prepared and evaluated in MM at preclinical and clinical levels. A summary overview of molecular functional imaging in MM is provided, and commonly used radiotracers are characterized.
Collapse
Affiliation(s)
- Shaojuan Zhang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jingjie Shang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Weijian Ye
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Tianming Zhao
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Hui Zeng
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- *Correspondence: Hui Zeng, ; Lu Wang,
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
- *Correspondence: Hui Zeng, ; Lu Wang,
| |
Collapse
|
11
|
Thomas E, Mathieu C, Moreno‐Gaona P, Mittelheisser V, Lux F, Tillement O, Pivot X, Ghoroghchian PP, Detappe A. Anti-BCMA Immuno-NanoPET Radiotracers for Improved Detection of Multiple Myeloma. Adv Healthc Mater 2022; 11:e2101565. [PMID: 34710281 PMCID: PMC11469021 DOI: 10.1002/adhm.202101565] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/09/2021] [Indexed: 11/07/2022]
Abstract
Current clinical imaging modalities for the sensitive and specific detection of multiple myeloma (MM) rely on nonspecific imaging contrast agents based on gadolinium chelates for magnetic resonance imaging (MRI) or for 18 F-FDG-directed and combined positron emission tomography (PET) and computed tomography (CT) scans. These tracers are not, however, able to detect minute plasma cell populations in the tumor niche, leading to false negative results. Here, a novel PET-based anti-BCMA nanoplatform labeled with 64 Cu is developed to improve the monitoring of these cells in both the spine and femur and to compare its sensitivity and specificity to more conventional immunoPET (64 Cu labeled anti-BCMA antibody) and passively targeted PET radiotracers (64 CuCl2 and 18 F-FDG). This proof-of-concept preclinical study confirmed that by conjugating up to four times more radioisotopes per antibody with the immuno-nanoPET platform, an improvement in the sensitivity and in the specificity of PET to detect tumor cells in an orthotopic model of MM is observed when compared to the traditional immunoPET approach. It is anticipated that when combined with tumor biopsy, this immuno-nanoPET platform may improve the management of patients with MM.
Collapse
Affiliation(s)
- Eloise Thomas
- LAGEPP Université Claude Bernard Lyon 1CNRS UMR5007VilleurbanneFrance
| | - Clélia Mathieu
- Université Paris‐SaclayCNRS UMR 8612Institut Galien Paris‐SaclayFrance
| | | | | | - François Lux
- Institut Lumière‐MatièreUniversité Claude Bernard Lyon 1CNRS UMR5306VilleurbanneFrance
- Institut Universitaire de France (IUF)ParisFrance
| | - Olivier Tillement
- Institut Lumière‐MatièreUniversité Claude Bernard Lyon 1CNRS UMR5306VilleurbanneFrance
| | - Xavier Pivot
- Institut de Cancérologie Strasbourg Europe (ICANS)StrasbourgFrance
| | - Paiman Peter Ghoroghchian
- David H Koch Institute for Integrative Cancer ResearchMITCambridgeMAUSA
- Dana Farber Cancer InstituteBostonMAUSA
| | - Alexandre Detappe
- Institut de Cancérologie Strasbourg Europe (ICANS)StrasbourgFrance
- Strasbourg Drug Discovery and Development Institute (IMS)StrasbourgFrance
| |
Collapse
|
12
|
Dun Y, Huang G, Liu J, Wei W. ImmunoPET imaging of hematological malignancies: From preclinical promise to clinical reality. Drug Discov Today 2021; 27:1196-1203. [PMID: 34838729 DOI: 10.1016/j.drudis.2021.11.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 10/22/2021] [Accepted: 11/18/2021] [Indexed: 12/23/2022]
Abstract
Immuno-positron emission tomography (immunoPET) imaging is a paradigm-shifting imaging technique for whole-body and all-lesion tumor detection, based on the combined specificity of tumor-targeting vectors [e.g., monoclonal antibodies (mAbs), nanobodies, and bispecific antibodies] and the sensitivity of PET imaging. By noninvasively, comprehensively, and serially revealing heterogeneous tumor antigen expression, immunoPET imaging is gradually improving the theranostic prospects for hematological malignancies. In this review, we summarize the available literature regarding immunoPET in imaging hematological malignancies. We also highlight the pros and cons of current conjugation strategies, and modular chemistry that can be leveraged to develop novel immunoPET probes for hematological malignancies. Lastly, we discuss the use of immunoPET imaging in guiding antibody drug development.
Collapse
Affiliation(s)
- Yiting Dun
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China.
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China.
| |
Collapse
|
13
|
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]
|
14
|
Liapis V, Tieu W, Wittwer NL, Gargett T, Evdokiou A, Takhar P, Rudd SE, Donnelly PS, Brown MP, Staudacher AH. Positron Emission Tomographic Imaging of Tumor Cell Death Using Zirconium-89-Labeled APOMAB® Following Cisplatin Chemotherapy in Lung and Ovarian Cancer Xenograft Models. Mol Imaging Biol 2021; 23:914-928. [PMID: 34231102 PMCID: PMC8578059 DOI: 10.1007/s11307-021-01620-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/28/2021] [Accepted: 05/26/2021] [Indexed: 02/02/2023]
Abstract
PURPOSE Early detection of tumor treatment responses represents an unmet clinical need with no approved noninvasive methods. DAB4, or its chimeric derivative, chDAB4 (APOMAB®) is an antibody that targets the Lupus associated antigen (La/SSB). La/SSB is over-expressed in malignancy and selectively targeted by chDAB4 in cancer cells dying from DNA-damaging treatment. Therefore, chDAB4 is a unique diagnostic tool that detects dead cancer cells and thus could distinguish between treatment responsive and nonresponsive patients. PROCEDURES In clinically relevant tumor models, mice bearing subcutaneous xenografts of human ovarian or lung cancer cell lines or intraperitoneal ovarian cancer xenografts were untreated or given chemotherapy followed 24h later by chDAB4 radiolabeled with [89Zr]ZrIV. Tumor responses were monitored using bioluminescence imaging and caliper measurements. [89Zr]Zr-chDAB4 uptake in tumor and normal tissues was measured using an Albira SI Positron-Emission Tomography (PET) imager and its biodistribution was measured using a Hidex gamma-counter. RESULTS Tumor uptake of [89Zr]Zr-chDAB4 was detected in untreated mice, and uptake significantly increased in both human lung and ovarian tumors after chemotherapy, but not in normal tissues. CONCLUSION Given that tumors, rather than normal tissues, were targeted after chemotherapy, these results support the clinical development of chDAB4 as a radiodiagnostic imaging agent and as a potential predictive marker of treatment response.
Collapse
Affiliation(s)
- Vasilios Liapis
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Level 9 University of South Australia Health Innovation Building, North Terrace, Adelaide, 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
| | - Nicole L Wittwer
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Level 9 University of South Australia Health Innovation Building, North Terrace, Adelaide, 5000, Australia
| | - Tessa Gargett
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Level 9 University of South Australia Health Innovation Building, North Terrace, Adelaide, 5000, Australia
| | - Andreas Evdokiou
- Discipline of Surgery, Breast Cancer Research Unit, Basil Hetzel Institute and Centre for Personalised Cancer Medicine, University of Adelaide, Woodville, SA, 5011, Australia
| | - Prab Takhar
- 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
| | - Michael P Brown
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Level 9 University of South Australia Health Innovation Building, North Terrace, Adelaide, 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, Level 9 University of South Australia Health Innovation Building, North Terrace, Adelaide, 5000, Australia
- School of Medicine, University of Adelaide, Adelaide, SA, 5000, Australia
| |
Collapse
|
15
|
Katal S, Maldonado A, Carrascoso J, Assadi M, Gholamrezanezhad A. Theranostic Agents in Musculoskeletal Disorders. PET Clin 2021; 16:441-448. [PMID: 34053587 DOI: 10.1016/j.cpet.2021.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Theranostic-based strategies, combining therapeutic and diagnostic properties of a single agent, have gained enormous attention in the past few years. Today, various multifunctional theranostic modalities have been examined, using different bioactive targeting, for the detection, quantifying, and monitoring of therapy response in different pathologies. Herein we review the newly emerging approaches in theranostic nanomedicine for the detection and therapy for musculoskeletal disorders to provide valuable insights for developing more efficient agents for clinical use. Some potential preclinical applications of radionuclide nanotheranostic agents are described in rheumatoid arthritis, osteoarthrosis, multiple myeloma, and neoplastic diseases.
Collapse
Affiliation(s)
- Sanaz Katal
- Department of Nuclear Medicine, Kowsar Hospital, Shiraz, Iran
| | - Antonio Maldonado
- Department of Nuclear Medicine, Quironsalud Madrid University Hospital, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Javier Carrascoso
- Department of Radiology, Quironsalud Madrid University Hospital, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Majid Assadi
- Department of Molecular Imaging and Radionuclide Therapy (MIRT), The Persian Gulf Nuclear Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Ali Gholamrezanezhad
- Department of Diagnostic Radiology, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA, USA.
| |
Collapse
|
16
|
Chomet M, van Dongen GAMS, Vugts DJ. State of the Art in Radiolabeling of Antibodies with Common and Uncommon Radiometals for Preclinical and Clinical Immuno-PET. Bioconjug Chem 2021; 32:1315-1330. [PMID: 33974403 PMCID: PMC8299458 DOI: 10.1021/acs.bioconjchem.1c00136] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
Inert
and stable radiolabeling of monoclonal antibodies (mAb),
antibody fragments, or antibody mimetics with radiometals is a prerequisite
for immuno-PET. While radiolabeling is preferably fast, mild, efficient,
and reproducible, especially when applied for human use in a current
Good Manufacturing Practice compliant way, it is crucial that the
obtained radioimmunoconjugate is stable and shows preserved immunoreactivity
and in vivo behavior. Radiometals and chelators have
extensively been evaluated to come to the most ideal radiometal–chelator
pair for each type of antibody derivative. Although PET imaging of
antibodies is a relatively recent tool, applications with 89Zr, 64Cu, and 68Ga have greatly increased in
recent years, especially in the clinical setting, while other less
common radionuclides such as 52Mn, 86Y, 66Ga, and 44Sc, but also 18F as in [18F]AlF are emerging promising candidates for the radiolabeling
of antibodies. This review presents a state of the art overview of
the practical aspects of radiolabeling of antibodies, ranging from
fast kinetic affibodies and nanobodies to slow kinetic intact mAbs.
Herein, we focus on the most common approach which consists of first
modification of the antibody with a chelator, and after eventual storage
of the premodified molecule, radiolabeling as a second step. Other
approaches are possible but have been excluded from this review. The
review includes recent and representative examples from the literature
highlighting which radiometal–chelator–antibody combinations
are the most successful for in vivo application.
Collapse
Affiliation(s)
- Marion Chomet
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam 1081 HV, The Netherlands
| | - Guus A M S van Dongen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam 1081 HV, The Netherlands
| | - Danielle J Vugts
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam 1081 HV, The Netherlands
| |
Collapse
|
17
|
Le Bihan T, Driver CHS, Ebenhan T, Le Bris N, Zeevaart JR, Tripier R. In Vivo Albumin-Binding of a C-Functionalized Cyclam Platform for 64 Cu-PET/CT Imaging in Breast Cancer Model. ChemMedChem 2020; 16:809-821. [PMID: 33191627 DOI: 10.1002/cmdc.202000800] [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: 10/13/2020] [Indexed: 11/06/2022]
Abstract
An improved glucose-chelator-albumin bioconjugate (GluCAB) derivative, GluCAB-2Mal , has been synthesized and studied for in vivo 64 Cu-PET/CT imaging in breast cancer mice models together with its first-generation analogue GluCAB-1Mal . The radioligand works on the principle of tumor targeting through the enhanced permeability and retention (EPR) effect with a supportive role played by glucose metabolism. [64 Cu]Cu-GluCAB-2Mal (99 % RCP) exhibited high serum stability with immediate binding to serum proteins. In vivo experiments for comparison between tumor targeting of [64 Cu]Cu-GluCAB-2Mal and previous-generation [64 Cu]Cu-GluCAB-1Mal encompassed microPET/CT imaging and biodistribution analysis in an allograft E0771 breast cancer mouse model. Tumor uptake of [64 Cu]Cu-GluCAB-2Mal was clearly evident with twice as much accumulation as compared to its predecessor and a tumor/muscle ratio of up to 5 after 24 h. Further comparison indicated a decrease in liver accumulation for [64 Cu]Cu-Glu-CAB-2Mal .
Collapse
Affiliation(s)
- Thomas Le Bihan
- UMR CNRS 6521 CEMCA, University of Brest, 6 avenue Le Gorgeu, CS93837, 29200, Brest, France
| | - Cathryn H S Driver
- South African Nuclear Energy Corporation Radiochemistry and NuMeRI PreClinical Imaging Facility, Elias Motsoaledi Street, R104 Pelindaba, North West, 0240, South Africa
| | - Thomas Ebenhan
- South African Nuclear Energy Corporation Radiochemistry and NuMeRI PreClinical Imaging Facility, Elias Motsoaledi Street, R104 Pelindaba, North West, 0240, South Africa
| | - Nathalie Le Bris
- UMR CNRS 6521 CEMCA, University of Brest, 6 avenue Le Gorgeu, CS93837, 29200, Brest, France
| | - Jan Rijn Zeevaart
- South African Nuclear Energy Corporation Radiochemistry and NuMeRI PreClinical Imaging Facility, Elias Motsoaledi Street, R104 Pelindaba, North West, 0240, South Africa
| | - Raphaël Tripier
- UMR CNRS 6521 CEMCA, University of Brest, 6 avenue Le Gorgeu, CS93837, 29200, Brest, France
| |
Collapse
|
18
|
Berdal M, Gouard S, Eychenne R, Marionneau-Lambot S, Croyal M, Faivre-Chauvet A, Chérel M, Gaschet J, Gestin JF, Guérard F. Investigation on the reactivity of nucleophilic radiohalogens with arylboronic acids in water: access to an efficient single-step method for the radioiodination and astatination of antibodies. Chem Sci 2020; 12:1458-1468. [PMID: 34163909 PMCID: PMC8179031 DOI: 10.1039/d0sc05191h] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Easy access to radioiodinated and 211At-labelled bio(macro)molecules is essential to develop new strategies in nuclear imaging and targeted radionuclide therapy of cancers. Yet, the labelling of complex molecules with heavy radiohalogens is often poorly effective due to the multiple steps and intermediate purifications needed. Herein, we investigate the potential of arylboron chemistry as an alternative approach for the late stage labelling of antibodies. The reactivity of a model precursor, 4-chlorobenzeneboronic acid (1) with nucleophilic iodine-125 and astatine-211 was at first investigated in aqueous conditions. In the presence of a copper(ii) catalyst and 1,10-phenanthroline, quantitative radiochemical yields (RCYs) were achieved within 30 minutes at room temperature. The optimum conditions were then applied to a CD138 targeting monoclonal antibody (mAb) that has previously been validated for imaging and therapy in a preclinical model of multiple myeloma. RCYs remained high (>80% for 125I-labelling and >95% for 211At-labelling), and the whole procedure led to increased specific activities within less time in comparison with previously reported methods. Biodistribution study in mice indicated that targeting properties of the radiolabelled mAb were well preserved, leading to a high tumour uptake in a CD138 expressing tumour model. The possibility of divergent synthesis from a common modified carrier protein demonstrated herein opens facilitated perspectives in radiotheranostic applications with the radioiodine/211At pairs. Overall, the possibility to develop radiolabelling kits offered by this procedure should facilitate its translation to clinical applications. The high reactivity of astatine and iodine in water with arylboronic acids provides access to an efficient single-step antibody radiolabelling.![]()
Collapse
Affiliation(s)
- Marion Berdal
- Université de Nantes, CNRS, Inserm, CRCINA F-44000 Nantes France
| | - Sébastien Gouard
- Université de Nantes, CNRS, Inserm, CRCINA F-44000 Nantes France
| | - Romain Eychenne
- Université de Nantes, CNRS, Inserm, CRCINA F-44000 Nantes France .,Arronax GIP Saint-Herblain France
| | - Séverine Marionneau-Lambot
- Université de Nantes, CNRS, Inserm, CRCINA F-44000 Nantes France .,Department of Nuclear Medicine, CHU Nantes Nantes France
| | - Mikaël Croyal
- CRNH-O, Mass Spectrometry Core Facility F-44000 Nantes France.,NUN, INRA, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O F-44000 Nantes France
| | - Alain Faivre-Chauvet
- Université de Nantes, CNRS, Inserm, CRCINA F-44000 Nantes France .,Department of Nuclear Medicine, CHU Nantes Nantes France
| | - Michel Chérel
- Université de Nantes, CNRS, Inserm, CRCINA F-44000 Nantes France .,ICO-René Gauducheau Saint-Herblain France
| | - Joëlle Gaschet
- Université de Nantes, CNRS, Inserm, CRCINA F-44000 Nantes France
| | | | - François Guérard
- Université de Nantes, CNRS, Inserm, CRCINA F-44000 Nantes France
| |
Collapse
|
19
|
Targeted-Alpha-Therapy Combining Astatine-211 and anti-CD138 Antibody in A Preclinical Syngeneic Mouse Model of Multiple Myeloma Minimal Residual Disease. Cancers (Basel) 2020; 12:cancers12092721. [PMID: 32971984 PMCID: PMC7564412 DOI: 10.3390/cancers12092721] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Multiple myeloma is a cancer that remains incurable. Among the many therapies under evaluation, antibodies can be used as vehicles to target and deliver toxic radiation to the tumour cells. Our objective was therefore to investigate the potential of targeted alpha therapy, combining an anti-CD138 mAb with astatine-211, to destroy the residual cells responsible for relapse. We have shown in a mouse model that mimics human disease, that destroying multiple myeloma cells is feasible with low toxicity by injecting an anti-CD138 mAb coupled with astatine-211. This approach could eradicate residual cells after initial treatment and thus prevent recurrence. Abstract Despite therapeutic progress in recent years with the introduction of targeted therapies (daratumumab, elotuzumab), multiple myeloma remains an incurable cancer. The question is therefore to investigate the potential of targeted alpha therapy, combining an anti-CD138 antibody with astatine-211, to destroy the residual cells that cause relapses. A preclinical syngeneic mouse model, consisting of IV injection of 1 million of 5T33 cells in a KaLwRij C57/BL6 mouse, was treated 10 days later with an anti-mCD138 antibody, called 9E7.4, radiolabeled with astatine-211. Four activities of the 211At-9E7.4 radioimmunoconjugate were tested in two independent experiments: 370 kBq (n = 16), 555 kBq (n = 10), 740 kBq (n = 17) and 1100 kBq (n = 6). An isotype control was also tested at 555 kBq (n = 10). Biodistribution, survival rate, hematological parameters, enzymatic hepatic toxicity, histological examination and organ dosimetry were considered. The survival median of untreated mice was 45 days after engraftment. While the activity of 1100 kBq was highly toxic, the activity of 740 kBq offered the best efficacy with 65% of overall survival 150 days after the treatment with no evident sign of toxicity. This work demonstrates the pertinence of treating minimal residual disease of multiple myeloma with an anti-CD138 antibody coupled to astatine-211.
Collapse
|
20
|
ImmunoPET in Multiple Myeloma-What? So What? Now What? Cancers (Basel) 2020; 12:cancers12061467. [PMID: 32512883 PMCID: PMC7352991 DOI: 10.3390/cancers12061467] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/30/2020] [Accepted: 06/01/2020] [Indexed: 12/11/2022] Open
Abstract
Despite constant progress over the past three decades, multiple myeloma (MM) is still an incurable disease, and the identification of new biomarkers to better select patients and adapt therapy is more relevant than ever. Recently, the introduction of therapeutic monoclonal antibodies (mAbs) (including direct-targeting mAbs and immune checkpoint inhibitors) appears to have changed the paradigm of MM management, emphasizing the opportunity to cure MM patients through an immunotherapeutic approach. In this context, immuno-positron emission tomography (immunoPET), combining the high sensitivity and resolution of a PET camera with the specificity of a radiolabelled mAb, holds the capability to cement this new treatment paradigm for MM patients. It has the potential to non-invasively monitor the distribution of therapeutic antibodies or directly monitor biomarkers on MM cells, and to allow direct observation of potential changes over time and in response to various therapeutic interventions. Tumor response could, in the future, be anticipated more effectively to provide individualized treatment plans tailored to patients according to their unique imaging signatures. This work explores the important role played by immunotherapeutics in the management of MM, and focuses on some of the challenges for this drug class and the significant interest of companion imaging agents such as immunoPET.
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
|
Ho M, Bianchi G, Anderson KC. Proteomics-inspired precision medicine for treating and understanding multiple myeloma. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020; 5:67-85. [PMID: 34414281 DOI: 10.1080/23808993.2020.1732205] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Introduction Remarkable progress in molecular characterization methods has led to significant improvements in how we manage multiple myeloma (MM). The introduction of novel therapies has led to significant improvements in overall survival over the past 10 years. However, MM remains incurable and treatment choice is largely based on outdated risk-adaptive strategies that do not factor in improved treatment outcomes in the context of modern therapies. Areas covered This review discusses current risk-adaptive strategies in MM and the clinical application of proteomics in the monitoring of treatment response, disease progression, and minimal residual disease (MRD). We also discuss promising biomarkers of disease progression, treatment response, and chemoresistance. Finally, we will discuss an immunomics-based approach to monoclonal antibody (mAb), vaccine, and CAR-T cell development. Expert opinion It is an exciting era in oncology with basic scientific knowledge translating in novel therapeutic approaches to improve patient outcomes. With the advent of effective immunotherapies and targeted therapies, it has become crucial to identify biomarkers to aid in the stratification of patients based on anticipated sensitivity to chemotherapy. As a paradigm of diseases highly dependent on protein homeostasis, multiple myeloma provides the perfect opportunity to investigate the use of proteomics to aid in precision medicine.
Collapse
Affiliation(s)
- Matthew Ho
- UCD School of Medicine, College of Health and Agricultural Sciences, University College Dublin, Dublin, Ireland
| | - Giada Bianchi
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kenneth C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|