1
|
Tunçel A, Maschauer S, Prante O, Yurt F. In Vitro Assessment of 177Lu-Labeled Trastuzumab-Targeted Mesoporous Carbon@Silica Nanostructure for the Treatment of HER2-Positive Breast Cancer. Pharmaceuticals (Basel) 2024; 17:732. [PMID: 38931400 PMCID: PMC11206869 DOI: 10.3390/ph17060732] [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: 05/10/2024] [Revised: 05/27/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024] Open
Abstract
This study assessed the effectiveness of a trastuzumab-targeted 177Lu-labeled mesoporous Carbon@Silica nanostructure (DOTA@TRA/MC@Si) for HER2-positive breast cancer treatment, focusing on its uptake, internalization, and efflux in breast cancer cells. The synthesized PEI-MC@Si nanocomposite was reacted with DOTA-NHS-ester, confirmed by the Arsenazo(III) assay. Following this, TRA was conjugated to the DOTA@PEI-MC@Si for targeting. DOTA@PEI-MC@Si and DOTA@TRA/MC@Si nanocomposites were labeled with 177Lu, and their efficacy was evaluated through in vitro radiolabeling experiments. According to the results, the DOTA@TRA/MC@Si nanocomposite was successfully labeled with 177Lu, yielding a radiochemical yield of 93.0 ± 2.4%. In vitro studies revealed a higher uptake of the [177Lu]Lu-DOTA@TRA/MC@Si nanocomposite in HER2-positive SK-BR-3 cells (44.0 ± 4.6% after 24 h) compared to MDA-MB-231 cells (21.0 ± 2.3%). The IC50 values for TRA-dependent uptake in the SK-BR-3 and BT-474 cells were 0.9 µM and 1.3 µM, respectively, indicating affinity toward HER-2 receptor-expressing cells. The lipophilic distribution coefficients of the radiolabeled nanocomposites were determined to be 1.7 ± 0.3 for [177Lu]Lu-DOTA@TRA/MC@Si and 1.5 ± 0.2 for [177Lu]Lu-DOTA@PEI-MC@Si, suggesting sufficient passive transport through the cell membrane and increased accumulation in target tissues. The [177Lu]Lu-DOTA@TRA/MC@Si nanocomposite showed an uptake into HER2-positive cell lines, marking a valuable step toward the development of a nanoparticle-based therapeutic agent for an improved treatment strategy for HER2-positive breast cancer.
Collapse
Affiliation(s)
- Ayça Tunçel
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova 35100, Turkey;
| | - Simone Maschauer
- Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Ulmenweg 18, D-91054 Erlangen, Germany;
| | - Olaf Prante
- Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Ulmenweg 18, D-91054 Erlangen, Germany;
| | - Fatma Yurt
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova 35100, Turkey;
| |
Collapse
|
2
|
Dhaouadi S, Bouhaouala-Zahar B, Orend G. Tenascin-C targeting strategies in cancer. Matrix Biol 2024; 130:1-19. [PMID: 38642843 DOI: 10.1016/j.matbio.2024.04.002] [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: 12/20/2023] [Revised: 04/13/2024] [Accepted: 04/14/2024] [Indexed: 04/22/2024]
Abstract
Tenascin-C (TNC) is a matricellular and multimodular glycoprotein highly expressed under pathological conditions, especially in cancer and chronic inflammatory diseases. Since a long time TNC is considered as a promising target for diagnostic and therapeutic approaches in anti-cancer treatments and was already extensively targeted in clinical trials on cancer patients. This review provides an overview of the current most advanced strategies used for TNC detection and anti-TNC theranostic approaches including some advanced clinical strategies. We also discuss novel treatment protocols, where targeting immune modulating functions of TNC could be center stage.
Collapse
Affiliation(s)
- Sayda Dhaouadi
- Laboratoire des Venins et Biomolécules Thérapeutiques, Institut Pasteur de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Balkiss Bouhaouala-Zahar
- Laboratoire des Venins et Biomolécules Thérapeutiques, Institut Pasteur de Tunis, Université Tunis El Manar, Tunis, Tunisia; Faculté de Médecine de Tunis, Université Tunis el Manar, Tunis, Tunisia
| | - Gertraud Orend
- INSERM U1109, The Tumor Microenvironment laboratory, Université Strasbourg, Hôpital Civil, Institut d'Hématologie et d'Immunologie, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
| |
Collapse
|
3
|
Chen W, Wu Y, Wang J, Yu W, Shen X, Zhao K, Liang B, Hu X, Wang S, Jiang H, Liu X, Zhang M, Xing X, Wang C, Xing D. Clinical advances in TNC delivery vectors and their conjugate agents. Pharmacol Ther 2024; 253:108577. [PMID: 38081519 DOI: 10.1016/j.pharmthera.2023.108577] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/02/2023] [Accepted: 12/04/2023] [Indexed: 12/17/2023]
Abstract
Tenascin C (TNC), a glycoprotein that is abundant in the tumor extracellular matrix (ECM), is strongly overexpressed in tumor tissues but virtually undetectable in most normal tissues. Many TNC antibodies, peptides, aptamers, and nanobodies have been investigated as delivery vectors, including 20A1, α-A2, α-A3, α-IIIB, α-D, BC-2, BC-4 BC-8, 81C6, ch81C6, F16, FHK, Ft, Ft-NP, G11, G11-iRGD, GBI-10, 19H12, J1/TN1, J1/TN2, J1/TN3, J1/TN4, J1/TN5, NJT3, NJT4, NJT6, P12, PL1, PL3, R6N, SMART, ST2146, ST2485, TN11, TN12, TNFnA1A2-Fc, TNfnA1D-Fc, TNfnBD-Fc, TNFnCD-Fc, TNfnD6-Fc, TNfn78-Fc, TTA1, TTA1.1, and TTA1.2. In particular, BC-2, BC-4, 81C6, ch81C6, F16, FHK, G11, PL1, PL3, R6N, ST2146, TN11, and TN12 have been tested in human tissues. G11-iRGD and simultaneous multiple aptamers and arginine-glycine-aspartic acid (RGD) targeting (SMART) may be assessed in clinical trials because G11, iRGD and AS1411 (SMART components) are already in clinical trials. Many TNC-conjugate agents, including antibody-drug conjugates (ADCs), antibody fragment-drug conjugates (FDCs), immune-stimulating antibody conjugates (ISACs), and radionuclide-drug conjugates (RDCs), have been investigated in preclinical and clinical trials. RDCs investigated in clinical trials include 111In-DTPA-BC-2, 131I-BC-2, 131I-BC-4, 90Y-BC4, 131I81C6, 131I-ch81C6, 211At-ch81C6, F16124I, 131I-tenatumomab, ST2146biot, FDC 131I-F16S1PF(ab')2, and ISAC F16IL2. ADCs (including FHK-SSL-Nav, FHK-NB-DOX, Ft-NP-PTX, and F16*-MMAE) and ISACs (IL12-R6N and 125I-G11-IL2) may enter clinical trials because they contain components of marketed treatments or agents that were investigated in previous clinical studies. This comprehensive review presents historical perspectives on clinical advances in TNC-conjugate agents to provide timely information to facilitate tumor-targeting drug development using TNC.
Collapse
Affiliation(s)
- Wujun Chen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China
| | - Yudong Wu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China
| | - Jie Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China
| | - Wanpeng Yu
- Qingdao Medical College, Qingdao University, Qingdao, Shandong 266071, China
| | - Xin Shen
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, Shandong 266042, China
| | - Kai Zhao
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China; Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Bing Liang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China
| | - Xiaokun Hu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China; Interventional Medicine Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Shuai Wang
- Department of Radiotherapy, Affiliated Hospital of Weifang Medical University, Key Laboratory of Precision Radiation Therapy for Tumors in Weifang City, School of Medical Imaging, Weifang Medical University, Weifang, Shandong 261031, China
| | - Hongfei Jiang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China
| | - Xinlin Liu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China
| | - Miao Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China
| | - Xiaohui Xing
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, China.
| | - Chao Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China.
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266000, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
4
|
Egorova BV, Fedorova OA, Kalmykov SN. Cationic radionuclides and ligands for targeted therapeutic radiopharmaceuticals. RUSSIAN CHEMICAL REVIEWS 2019. [DOI: 10.1070/rcr4890] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
This review considers the already used and potential α- and β-emitting cationic radionuclides for targeted radionuclide therapy. Recent results of laboratory, preclinical and clinical applications of these radionuclides are discussed. As opposed to β-emitters, which are already used in nuclear medicine, α-emitters involved in targeted radiopharmaceuticals were subjected to clinical trials only recently and were found to be therapeutically effective. The review summarizes recent trends in the development of ligands as components of radiopharmaceuticals addressing specific features of short-lived cationic radionuclides applied in medicine. Despite a steadily growing number of chelating ligands, 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) and diethylenetriaminepentaacetic acid (DTPA) remain the most widely used agents in nuclear medicine. The drawbacks of these compounds restrict the application of radionuclides in medicine. Variations in the macrocycle size, the introduction and modification of substituents can significantly improve the chelating ability of ligands, enhance stability of radionuclide complexes with these ligands and eliminate the influence of ligands on the affinity of biological targeting vectors.
The bibliography includes 189 references.
Collapse
|
5
|
Ma W, Wang X, Liu W, Ma H, Su Y, Yang Y, Liu N, Wang Y, Yang G. A Theoretical Model for Predicting and Optimizing In Vitro Screening of Potential Targeted Alpha-Particle Therapy Drugs. Radiat Res 2019; 191:475-482. [PMID: 30830838 DOI: 10.1667/rr15297.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
One highly promising approach to cancer treatment, especially for tumors that have undergone micrometastasis, is targeted alpha-particle therapy (TAT). However, the development of a TAT drug has been impeded due to numerous unsuccessful attempts to establish effective in vitro screening methods. The goal of this study was to construct a model to predict and optimize in vitro screening of potential TAT drugs. Based on mean field hypothesis, microdosimetry and the classic linear-quadratic equation, a novel model was built, which can predict our own in vitro experiments and replicate published data from others. Interestingly, this model can also be used to quickly optimize several key parameters in in vitro screening of potential TAT drugs, instructing the optimal combinations of the expression level of antigen, the binding affinity of antibody and drug antibody ratio, as well as others. In addition, to conveniently evaluate the therapeutic benefit of different drugs, a simple but universal parameter, the death ratio, is proposed. To our knowledge, this is the first model that can predict and guide the optimization of in vitro potential targeted alpha-particle therapy drug screening, which may then accelerate the development of potential targeted alpha-particle therapy drugs dramatically.
Collapse
Affiliation(s)
- Wenzong Ma
- a State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, P. R. China
| | - Xudong Wang
- a State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, P. R. China
| | - Weihao Liu
- b Key Laboratory of Radiation Physics and Technology, Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, P. R. China
| | - Huan Ma
- b Key Laboratory of Radiation Physics and Technology, Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, P. R. China
| | - Yue Su
- a State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, P. R. China
| | - Yuanyou Yang
- b Key Laboratory of Radiation Physics and Technology, Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, P. R. China
| | - Ning Liu
- b Key Laboratory of Radiation Physics and Technology, Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, P. R. China
| | - Yugang Wang
- a State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, P. R. China
| | - Gen Yang
- a State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, P. R. China
| |
Collapse
|
6
|
Basaco T, Pektor S, Bermudez JM, Meneses N, Heller M, Galván JA, Boligán KF, Schürch S, von Gunten S, Türler A, Miederer M. Evaluation of Radiolabeled Girentuximab In Vitro and In Vivo. Pharmaceuticals (Basel) 2018; 11:E132. [PMID: 30487460 PMCID: PMC6316122 DOI: 10.3390/ph11040132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 11/16/2022] Open
Abstract
Girentuximab (cG250) targets carbonic anhydrase IX (CAIX), a protein which is expressed on the surface of most renal cancer cells (RCCs). cG250 labeled with 177Lu has been used in clinical trials for radioimmunotherapy (RIT) of RCCs. In this work, an extensive characterization of the immunoconjugates allowed optimization of the labeling conditions with 177Lu while maintaining immunoreactivity of cG250, which was then investigated in in vitro and in vivo experiments. cG250 was conjugated with S-2-(4-isothiocyanatobenzyl)-1,4,7,10-tetraazacyclododecane tetraacetic acid (DOTA(SCN)) by using incubation times between 30 and 90 min and characterized by mass spectrometry. Immunoconjugates with five to ten DOTA(SCN) molecules per cG250 molecule were obtained. Conjugates with ratios less than six DOTA(SCN)/cG250 had higher in vitro antigen affinity, both pre- and postlabeling with 177Lu. Radiochemical stability increased, in the presence of sodium ascorbate, which prevents radiolysis. The immunoreactivity of the radiolabeled cG250 tested by specific binding to SK-RC-52 cells decreased when the DOTA content per conjugate increased. The in vivo tumor uptake was < 10% ID/g and independent of the total amount of protein in the range between 5 and 100 µg cG250 per animal. Low tumor uptake was found to be due to significant necrotic areas and heterogeneous CAIX expression. In addition, low vascularity indicated relatively poor accessibility of the CAIX target.
Collapse
Affiliation(s)
- Tais Basaco
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
- Laboratory of Radiochemistry, Paul Scherrer Institute (PSI), 5232 Villigen PSI, Switzerland.
| | - Stefanie Pektor
- Clinic for Nuclear Medicine, University Medical Center Mainz, 55131 Mainz, Germany.
| | - Josue M Bermudez
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
| | - Niurka Meneses
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
| | - Manfred Heller
- Department for Biomedical Research (DBMR), University of Bern, 3010 Bern, Switzerland.
| | - José A Galván
- Institute of Pathology, University of Bern, 3010 Bern, Switzerland.
| | - Kayluz F Boligán
- Institute of Pharmacology (PKI), University of Bern, 3010 Bern, Switzerland.
| | - Stefan Schürch
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
| | - Stephan von Gunten
- Institute of Pharmacology (PKI), University of Bern, 3010 Bern, Switzerland.
| | - Andreas Türler
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
| | - Matthias Miederer
- Clinic for Nuclear Medicine, University Medical Center Mainz, 55131 Mainz, Germany.
| |
Collapse
|
7
|
Cabello G, Nwoko KC, Mingarelli M, McLaughlin AC, Trembleau L, Feldmann J, Cuesta A, Smith TA. Physicochemical Tools: Toward a Detailed Understanding of the Architecture of Targeted Radiotherapy Nanoparticles. ACS APPLIED BIO MATERIALS 2018; 1:1639-1646. [DOI: 10.1021/acsabm.8b00476] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Gema Cabello
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, United Kingdom
| | - Kenneth C. Nwoko
- Department of Chemistry, School of Natural and Computing Sciences, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom
| | - Marco Mingarelli
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, United Kingdom
| | - Abbie C. McLaughlin
- Department of Chemistry, School of Natural and Computing Sciences, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom
| | - Laurent Trembleau
- Department of Chemistry, School of Natural and Computing Sciences, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom
| | - Joerg Feldmann
- Department of Chemistry, School of Natural and Computing Sciences, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom
| | - Angel Cuesta
- Department of Chemistry, School of Natural and Computing Sciences, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom
| | - Tim A.D. Smith
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, United Kingdom
| |
Collapse
|
8
|
Egorova BV, Oshchepkov MS, Fedorov YV, Fedorova OA, Budylin GS, Shirshin EA, Kalmykov SN. Complexation of Bi3+, Ac3+, Y3+, Lu3+, La3+ and Eu3+ with benzo-diaza-crown ether with carboxylic pendant arms. RADIOCHIM ACTA 2016. [DOI: 10.1515/ract-2015-2560] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Polyaminopolycarboxylates are attractive ligands for binding cationic radionuclides for synthesis of radiopharmaceuticals with target delivery to tumor cells. Nowadays beta emitting Y−90 and Lu−177 are used as therapeutic agents, while Ac−225 and Bi−213 are considered as perspective for alpha therapy. In the present study new data on complexation of Y3+, Lu3+, Ac3+ and Bi3+ with 2,2’-(15-formyl-2,3,5,6,8,9,11,12-octahydrobenzo [b][1,4,10,7,13]trioxadiazacyclopentadecine-4,10- diyl)diacetic acid are presented. For ligand and complexes characterization potentiometric titration, solvent extraction, chromatography and solubility techniques were applied. The highest values of stability constants within the range of log K = 5.8 – 7.5 were found for Ac3+ and REE. Fast complex formation is established which is beneficial for practical applications in radiopharmaceutical synthesis.
Collapse
Affiliation(s)
- Bayirta V. Egorova
- Lomonosov Moscow State University , Chemistry Department , 119991 Leninskie Gory, 1/3, Moscow, Russian Federation
| | - Maxim S. Oshchepkov
- Mendeleev University of Chemistry and Technology of Russia , 125047 Miusskaya sqr., 9, Moscow, Russian Federation
- A. N. Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, 119991 Vavilova, 28, GSP-1, Moscow, Russian Federation
| | - Yury V. Fedorov
- Lomonosov Moscow State University , Chemistry Department , 119991 Leninskie Gory, 1/3, Moscow, Russian Federation
- A. N. Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, 119991 Vavilova, 28, GSP-1, Moscow, Russian Federation
| | - Olga A. Fedorova
- Mendeleev University of Chemistry and Technology of Russia , 125047 Miusskaya sqr., 9, Moscow, Russian Federation
- A. N. Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, 119991 Vavilova, 28, GSP-1, Moscow, Russian Federation
| | - Gleb S. Budylin
- Lomonosov Moscow State University , Department of Physics, 119991 Leninskie Gory, 1/2, Moscow, Russian Federation
| | - Evgeny A. Shirshin
- Lomonosov Moscow State University , Department of Physics, 119991 Leninskie Gory, 1/2, Moscow, Russian Federation
| | - Stepan N. Kalmykov
- Lomonosov Moscow State University , Chemistry Department , 119991 Leninskie Gory, 1/3, Moscow, Russian Federation
- National Research Center “Kurchatov Institute”, 123182 Akademika Kurchatova sqr., 1, Moscow, Russian Federation
| |
Collapse
|
9
|
Rasaneh S, Rajabi H, Johari Daha F. Activity estimation in radioimmunotherapy using magnetic nanoparticles. Chin J Cancer Res 2015; 27:203-8. [PMID: 25937783 DOI: 10.3978/j.issn.1000-9604.2015.03.06] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 12/11/2014] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE Estimation of activity accumulated in tumor and organs is very important in predicting the response of radiopharmaceuticals treatment. In this study, we synthesized (177)Lutetium ((177)Lu)-trastuzumab-iron oxide nanoparticles as a double radiopharmaceutical agent for treatment and better estimation of organ activity in a new way by magnetic resonance imaging (MRI). METHODS (177)Lu-trastuzumab-iron oxide nanoparticles were synthesized and all the quality control tests such as labeling yield, nanoparticle size determination, stability in buffer and blood serum up to 4 d, immunoreactivity and biodistribution in normal mice were determined. In mice bearing breast tumor, liver and tumor activities were calculated with three methods: single photon emission computed tomography (SPECT), MRI and organ extraction, which were compared with each other. RESULTS The good results of quality control tests (labeling yield: 61%±2%, mean nanoparticle hydrodynamic size: 41±15 nm, stability in buffer: 86%±5%, stability in blood serum: 80%±3%, immunoreactivity: 80%±2%) indicated that (177)Lu-trastuzumab-iron oxide nanoparticles could be used as a double radiopharmaceutical agent in mice bearing tumor. Results showed that (177)Lu-trastuzumab-iron oxide nanoparticles with MRI had the ability to measure organ activities more accurate than SPECT. CONCLUSIONS Co-conjugating radiopharmaceutical to MRI contrast agents such as iron oxide nanoparticles may be a good way for better dosimetry in nuclear medicine treatment.
Collapse
Affiliation(s)
- Samira Rasaneh
- 1 Department of Radioisotope, Nuclear science and Technology Research Institute, Tehran 14115-331, Iran ; 2 Department of Medical Physics, School of Medical Sciences, Tarbiat Modares University, Tehran 14115-331, Iran
| | - Hossein Rajabi
- 1 Department of Radioisotope, Nuclear science and Technology Research Institute, Tehran 14115-331, Iran ; 2 Department of Medical Physics, School of Medical Sciences, Tarbiat Modares University, Tehran 14115-331, Iran
| | - Fariba Johari Daha
- 1 Department of Radioisotope, Nuclear science and Technology Research Institute, Tehran 14115-331, Iran ; 2 Department of Medical Physics, School of Medical Sciences, Tarbiat Modares University, Tehran 14115-331, Iran
| |
Collapse
|
10
|
Banerjee S, Pillai MRA, Knapp FFR. Lutetium-177 therapeutic radiopharmaceuticals: linking chemistry, radiochemistry, and practical applications. Chem Rev 2015; 115:2934-74. [PMID: 25865818 DOI: 10.1021/cr500171e] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Sharmila Banerjee
- Radiopharmaceuticals Chemistry Section, Bhabha Atomic Research Centre (BARC), Mumbai 400 085, India.,Molecular Group of Companies, Puthuvype, Ernakulam, Kerala 682 508, India.,Medical Radioisotope Program, Oak Ridge National Laboratory (ORNL), P.O. Box 2008, 1 Bethel Valley Road, Oak Ridge, Tennessee 37830-6229, United States
| | - M R A Pillai
- Radiopharmaceuticals Chemistry Section, Bhabha Atomic Research Centre (BARC), Mumbai 400 085, India.,Molecular Group of Companies, Puthuvype, Ernakulam, Kerala 682 508, India.,Medical Radioisotope Program, Oak Ridge National Laboratory (ORNL), P.O. Box 2008, 1 Bethel Valley Road, Oak Ridge, Tennessee 37830-6229, United States
| | - F F Russ Knapp
- Radiopharmaceuticals Chemistry Section, Bhabha Atomic Research Centre (BARC), Mumbai 400 085, India.,Molecular Group of Companies, Puthuvype, Ernakulam, Kerala 682 508, India.,Medical Radioisotope Program, Oak Ridge National Laboratory (ORNL), P.O. Box 2008, 1 Bethel Valley Road, Oak Ridge, Tennessee 37830-6229, United States
| |
Collapse
|
11
|
Physical optimization of production by deuteron irradiation of high specific activity 177gLu suitable for radioimmunotherapy. Nucl Med Biol 2014; 41:407-9. [DOI: 10.1016/j.nucmedbio.2014.02.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 02/20/2014] [Accepted: 02/21/2014] [Indexed: 11/18/2022]
|
12
|
Hill A, Breyer S, Geissler S, Mier W, Haberkorn U, Weigandt M, Mäder K. How do in-vitro release profiles of nanosuspensions from Alzet® pumps correspond to the in-vivo situation? A case study on radiolabeled fenofibrate. J Control Release 2013; 168:77-87. [PMID: 23499716 DOI: 10.1016/j.jconrel.2013.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 03/02/2013] [Accepted: 03/05/2013] [Indexed: 02/02/2023]
Abstract
In research and development sufficiently high and constant plasma levels of drug candidates are often requested, but simple solutions of hydrophobic drugs delivered from the commonly used micro-osmotic pumps cannot meet these demands. Nanosuspensions released from implanted osmotic devices can be a strategy to overcome this challenge but little is known about their pharmacokinetic behavior after subcutaneous application. In the current study, four different nanosuspension formulations containing iodinated fenofibrate were prepared, physicochemically characterized and investigated concerning their in-vitro release kinetics from osmotic pumps. One nanosuspension of lower viscosity exhibited thereby an unexpectedly first order release kinetics, whereas the higher viscous counterpart was released in the expected zero-order manner. To assess the relation of the in-vitro release kinetics to the in-vivo fate of nanosuspensions, various [(131)I] iodinated fenofibrate formulations were subcutaneously applied to mice. The biodistribution was followed by means of γ-scintigraphy and γ-scintillation. Two different nanosuspensions released from osmotic pumps were compared to bolus injections of a nanosuspension and an organic drug solution. The distribution and elimination of the bolus injected drug solution were almost completed within 48h. In contrast, a long lasting (>1week) depot at the injection site was formed by the bolus injected nanosuspension. Ex vivo examination of the organs showed a sustained, but exponential decrease of the radiolabel concentration. More constant drug levels in the organs were achieved within the nanosuspensions released from osmotic pumps. The organ levels of [(131)I] labeled fenofibrate were found to be more constant in case of the pump with the higher viscous nanosuspension in contrast to the lower viscous counterpart. However, the very different release profiles of the lower and higher viscous nanosuspension observed in-vitro were not observed in-vivo, as both pumps showed zero order release. In conclusion, nanosuspensions of poorly soluble compounds released from subcutaneously implanted osmotic pumps can be a suitable approach in pharmacokinetic studies. Although the in-vivo release of nanosuspensions differed in the expected release profile from the in-vitro test results, these in-vitro release tests present a valuable tool for the pre-selection of suitable nanosuspension candidates.
Collapse
Affiliation(s)
- A Hill
- Department of Pharmacy, Institute of Pharmaceutical Technology and Biopharmacy, Martin-Luther-University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle/Saale, Germany
| | | | | | | | | | | | | |
Collapse
|
13
|
Evaluation of strained alkynes for Cu-free click reaction in live mice. Nucl Med Biol 2013; 40:415-23. [PMID: 23380519 DOI: 10.1016/j.nucmedbio.2012.12.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 11/23/2012] [Accepted: 12/10/2012] [Indexed: 01/15/2023]
Abstract
INTRODUCTION We report on our evaluation of the strain-promoted cyclooctyne-azide cycloaddition reaction for use in tumor pretargeting, comprising a side-by-side comparison of probes 1-3 bearing three distinct cyclooctyne moieties based respectively on the 1st and 2nd generation difluorinated cyclooctyne and the 1st generation dibenzocyclooctyne. METHODS The probes were synthesized and labeled with (177)Lu with high yields. The probe stability and reactivity towards azides were evaluated in PBS and mouse serum, and their blood clearance, biodistribution and in vivo reactivity were evaluated in tumor-free mice. RESULTS In serum the three probes exhibited sufficient stability for a pretargeting application with half-lives of 12-19h. In PBS, probes 2 and 3 were more reactive towards azido-conjugated Rituximab (Rtx-N3) than 1, but in contrast to 1, their reactivity decreased in mouse serum and mouse serum albumin solutions, as a result of covalent and non-covalent interactions with albumin. Biodistribution data confirmed the interactions with serum proteins in circulation: (177)Lu-1 showed a fast elimination from blood (t1/2,β = 0.31h), while (177)Lu-2 and (177)Lu-3 were retained in blood for longer periods of time (t1/2,β = 1.08 and 3.58h, respectively). Dual isotope biodistribution experiments assessing the reaction between (125)I-Rtx-N3 and (177)Lu-1-3 in circulation in mice showed a very limited retention of 2 and 3 in blood rich organs, indicating a minimal reactivity, while no such retention was observed for 1. CONCLUSION The low reactivity of the studied cyclooctynes, and their serum interactions preclude their use at the low in vivo concentrations typical for pretargeting applications.
Collapse
|
14
|
D'Huyvetter M, Aerts A, Xavier C, Vaneycken I, Devoogdt N, Gijs M, Impens N, Baatout S, Ponsard B, Muyldermans S, Caveliers V, Lahoutte T. Development of 177Lu-nanobodies for radioimmunotherapy of HER2-positive breast cancer: evaluation of different bifunctional chelators. CONTRAST MEDIA & MOLECULAR IMAGING 2012; 7:254-64. [PMID: 22434639 DOI: 10.1002/cmmi.491] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Nanobodies show favourable pharmacokinetic characteristics for tumor targeting, including high tumor-to-background-ratios. Labelled with a therapeutic radionuclide, nanobodies could be used as an adjuvant treatment option for HER2-overexpressing minimal residual disease. The therapeutic radionuclide Lutetium-177 is linked to the nanobody using a bifunctional chelator. The choice of the bifunctional chelator could affect the in vivo behaviour of the radiolabeled nanobody. Consequently, we compared four different bifunctional chelators - p-SCN-Bn-DOTA, DOTA-NHS-ester, CHX-A"-DTPA or 1B4M-DTPA - in order to select the optimal chemical link between Lutetium-177 and a HER2 targeting nanobody. MS results revealed different degrees of chelator-conjugation. High stability in time was observed, together with nanomolar affinities on HER2-expressing tumor cells. Ex vivo biodistributions as well as SPECT/micro-CT analyses showed high activities in tumors expressing medium HER2 levels with low background activity except for the kidneys. The 1B4M-DTPA-coupled conjugate was further evaluated in a high HER2-expressing tumor model. Here, tumor uptake values of 5.99 ± 0.63, 5.12 ± 0.17, 2.83 ± 0.36 and 2.47 ± 0.38 %IA/g were obtained at 1, 3, 24 and 48h p.i., which coincided with exceptionally low background values, except for the kidneys, and unprecedented tumor-to-background ratios. No specific binding was observed in a HER2-negative model. In conclusion, the in-house developed anti-HER2 nanobody 2Rs15dHIS can be successfully labeled with (177) Lu using different bifunctional chelators. Both macrocyclic and acyclic chelators show high stability in time. High specific tumor uptake combined with the lowest background uptake was measured using the 1B4M-DTPA-based conjugate.
Collapse
Affiliation(s)
- Matthias D'Huyvetter
- Radiobiology Unit, Molecular and Cellular Biology Expert Group, Belgian Nuclear Research Center (SCK•CEN), Mol, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Hall MA, Aldrich MB, Azhdarinia A, Lachance PA, Robinson H, Hazen A, Haviland DL, Sevick-Muraca EM. Quantifying multimodal contrast agent biological activity using near-infrared flow cytometry. CONTRAST MEDIA & MOLECULAR IMAGING 2012; 7:338-45. [DOI: 10.1002/cmmi.502] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Mary A. Hall
- Center for Molecular Imaging; The Brown Foundation Institute of Molecular Medicine at The University of Texas Health Science Center; Houston; TX; 77030; USA
| | - Melissa B. Aldrich
- Center for Molecular Imaging; The Brown Foundation Institute of Molecular Medicine at The University of Texas Health Science Center; Houston; TX; 77030; USA
| | - Ali Azhdarinia
- Center for Molecular Imaging; The Brown Foundation Institute of Molecular Medicine at The University of Texas Health Science Center; Houston; TX; 77030; USA
| | - Pier-Anne Lachance
- Center for Molecular Imaging; The Brown Foundation Institute of Molecular Medicine at The University of Texas Health Science Center; Houston; TX; 77030; USA
| | - Holly Robinson
- Center for Molecular Imaging; The Brown Foundation Institute of Molecular Medicine at The University of Texas Health Science Center; Houston; TX; 77030; USA
| | - Amy Hazen
- Flow Cytometry Center; The Brown Foundation Institute of Molecular Medicine at The University of Texas Health Science Center; Houston; TX; 77030; USA
| | - David L. Haviland
- Flow Cytometry Center; The Brown Foundation Institute of Molecular Medicine at The University of Texas Health Science Center; Houston; TX; 77030; USA
| | - Eva M. Sevick-Muraca
- Center for Molecular Imaging; The Brown Foundation Institute of Molecular Medicine at The University of Texas Health Science Center; Houston; TX; 77030; USA
| |
Collapse
|
16
|
Vera DRB, Eigner S, Beran M, Henke KE, Laznickova A, Laznicek M, Melichar F, Chinol M. Preclinical evaluation of (177)lu-nimotuzumab: a potential tool for radioimmunotherapy of epidermal growth factor receptor-overexpressing tumors. Cancer Biother Radiopharm 2011; 26:287-97. [PMID: 21711096 DOI: 10.1089/cbr.2010.0916] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The humanized monoclonal antibody Nimotuzumab (h-R3) has demonstrated an exceptional and better clinical profile than other monoclonal antibodies for immunotherapy of epidermal growth factor receptor-overexpressing tumors. This work deals with the preparation and radiolabeling optimization of (177)Lu-Nimotuzumab and their preclinical evaluation. METHODS Nimotuzumab was conjugated with S-2-(4-isothiocyanatobenzyl)-1,4,7,10-tetraazacyclododecane tetraacetic acid (p-SCN-Bn-DOTA), testing different molar ratios. The immunoconjugates were characterized. The radiolabeling with (177)Lu was optimized. Radioimmunoconjugates stability was tested in 2-[bis[2-[bis(carboxymethyl)amino]ethyl]amino]acetic acid (DTPA) excess and human serum. In vitro studies were performed in tumor model cell lines. Receptor-specific binding was tested by competitive inhibition. (177)Lu-Nimotuzumab in vivo studies were conducted in healthy and xenograft animals. RESULTS Nimotuzumab conjugates were obtained with high purity. Radiolabeling yield and specific activities ranged from 63.6% to 94.5% and from 748 to 1142 MBq/mg, respectively. The stability in DTPA excess and human serum was 95.9% and 93.2% after 10 days, respectively. The radioimmunoconjugate showed specific receptor binding in tumor cell lines. Biodistribution in healthy animals showed the typical behavior of the immunoconjugates based on monoclonal antibodies. The study in xenografts mice demonstrated uptake of (177)Lu-Nimotuzumab in the tumor and reticuloendothelial organs. CONCLUSIONS (177)Lu-Nimotuzumab was obtained with high purity and specific activities under optimal conditions without significant loss in immunoreactivity and might be a potential radioimmunoconjugate for radioimmunotherapy of tumors with epidermal growth factor receptor overexpression.
Collapse
Affiliation(s)
- Denis Rolando Beckford Vera
- Radiopharmaceutical Department, Nuclear Physics Institute of the Academy of Sciences, Husinec-Rez, Czech Republic.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Hens M, Vaidyanathan G, Zhao XG, Bigner DD, Zalutsky MR. Anti-EGFRvIII monoclonal antibody armed with 177Lu: in vivo comparison of macrocyclic and acyclic ligands. Nucl Med Biol 2011; 37:741-50. [PMID: 20870149 DOI: 10.1016/j.nucmedbio.2010.04.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2009] [Revised: 03/23/2010] [Accepted: 04/04/2010] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Monoclonal antibody (mAb) L8A4 binds specifically to the epidermal growth factor receptor variant III (EGFRvIII) that is present on gliomas but not on normal tissues, and is internalized rapidly after receptor binding. Because of the short range of its β-emissions, labeling this mAb with (177)Lu would be an attractive approach for the treatment of residual tumor margins remaining after surgical debulking of brain tumors. MATERIALS AND METHODS L8A4 mAb was labeled with (177)Lu using the acyclic ligands [(R)-2-amino-3-(4-isothiocyanatophenyl)propyl]-trans-(S,S)-cyclohexane-1,2-diamine-pentaacetic acid (CHX-A″-DTPA) and 2-(4-isothiocyanatobenzyl)-6-methyldiethylene-triaminepentaacetic acid (1B4M-DTPA), and the macrocyclic ligands S-2-(4-isothiocyanatobenzyl)-1,4,7,10-tetraazacyclododecane-tetraacetic acid (C-DOTA) and α-(5-isothiocyanato-2-methoxyphenyl)-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (MeO-DOTA). Paired-label tissue distribution experiments were performed in athymic mice bearing subcutaneous EGFRvIII-expressing U87.ΔEGFR glioma xenografts over a period of 1 to 8 days to directly compare (177)Lu-labeled L8A4 to L8A4 labeled with (125)I using N-succinimidyl 4-guanidinomethyl-3-[(125)I]iodobenzoate ([(125)I]SGMIB). RESULTS Except with C-DOTA, tumor uptake for the (177)Lu-labeled mAb was significantly higher than the co-administered radioiodinated preparation; however, this was also the case for spleen, liver, bone and kidneys. Tumor/normal tissue ratios for (177)Lu-1B4M-DTPA-L8A4 and, to an even greater extent, (177)Lu-MeO-DOTA-L8A4 were higher than those for [(125)I]SGMIB-L8A4 in most other tissues. CONCLUSIONS Tumor and normal tissue distribution patterns for this anti-EGFRvIII mAb were dependent on the nature of the bifunctional chelate used for (177)Lu labeling. Optimal results were obtained with 1B4M-DTPA and MeO-DOTA, suggesting no clear advantage for acyclic vs. macrocyclic ligands for this application.
Collapse
Affiliation(s)
- Marc Hens
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
18
|
Manenti S, Groppi F, Gandini A, Gini L, Abbas K, Holzwarth U, Simonelli F, Bonardi M. Excitation function for deuteron induced nuclear reactions on natural ytterbium for production of high specific activity 177gLu in no-carrier-added form for metabolic radiotherapy. Appl Radiat Isot 2011; 69:37-45. [DOI: 10.1016/j.apradiso.2010.08.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2010] [Revised: 07/23/2010] [Accepted: 08/10/2010] [Indexed: 11/17/2022]
|
19
|
Rasaneh S, Rajabi H, Babaei MH, Daha FJ. 177Lu labeling of Herceptin and preclinical validation as a new radiopharmaceutical for radioimmunotherapy of breast cancer. Nucl Med Biol 2010; 37:949-55. [PMID: 21055626 DOI: 10.1016/j.nucmedbio.2010.07.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Revised: 05/30/2010] [Accepted: 07/01/2010] [Indexed: 11/27/2022]
Abstract
INTRODUCTION In the present study, Herceptin was labeled with lutetium-177 via DOTA, and the necessary preclinical quality control tests (in vitro and in vivo) were performed to evaluate its use as a radioimmunotherapy agent. MATERIAL AND METHODS Herceptin was conjugated to DOTA as a chelator in three different conjugation buffers (ammonium acetate, carbonate and HEPES buffer); each of the resulting conjugates was compared with respect to in vitro characteristics such as number of chelates per antibody, incorporated activity, immunoreactivity and in vitro stability in PBS buffer and blood serum. The biodistribution study and gamma camera imaging were performed in mice bearing breast tumors. To assess the therapeutic effects of (177)Lu-Herceptin, cytotoxicity was investigated for 7 days in a SKBr3 breast cancer cell line. RESULTS Carbonate buffer was the best conjugation buffer (number of chelates per antibody: 6; incorporated activity: 81%; immunoreactivity: 87%; buffer stability: 86%; serum stability: 81%, after 4 days). The efficient tumor uptake observed in the biodistribution studies was consistent with the gamma camera image results. At a concentration of 4 μg ml(-1), (177)Lu-Herceptin (surviving cells: 5 ± 0.6% of the total cells) of the total cells corresponded to an approximately eightfold increase in cytotoxicity in comparison to unmodified Herceptin (surviving cells: 43 ± 3.9%). CONCLUSION The new complex described herein could be considered for further evaluation in animals and potentially in humans as a radiopharmaceutical for use in the radioimmunotherapy of breast cancer. These results may be important for patients who cannot tolerate the therapeutic dosage of Herceptin currently used because of heart problems.
Collapse
Affiliation(s)
- Samira Rasaneh
- Department of Medical Physics, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14115-331, Tehran, Iran
| | | | | | | |
Collapse
|
20
|
|
21
|
Rasaneh S, Rajabi H, Babaei MH, Johari Daha F. Synthesis and biodistribution studiesof 177Lu-trastuzumab as a therapeutic agent in the breast cancer mice model. J Labelled Comp Radiopharm 2010. [DOI: 10.1002/jlcr.1780] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
22
|
Rasaneh S, Rajabi H, Hossein Babaei M, Johari Daha F. Toxicity of trastuzumab labeled 177Lu on MCF7 and SKBr3 cell lines. Appl Radiat Isot 2010; 68:1964-6. [PMID: 20537546 DOI: 10.1016/j.apradiso.2010.05.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2009] [Revised: 04/11/2010] [Accepted: 05/06/2010] [Indexed: 11/16/2022]
Abstract
In this study, we labeled trastuzumab with (177)Lu to synthesize a new radiopharmaceutical for therapy of breast cancer and at the first stage investigated its therapeutic effects on SKBr3 and MCF7 breast cancer cell lines. Trastuzumab-(177)Lu showed very good in-vitro characteristics such as high radiochemical purity (91+/-0.9%), good stability in PBS buffer (86+/-2.3%) and blood serum (81+/-2.7%) up to 96 h, appropriate immunoreactivity (85.4+/-1.1%) and high cytotoxicity in HER2 expression cells. 5 fold increase in toxicity of trastuzumab-(177)Lu was observed when compared with unlabeled trastuzumab on SKBr3 cells.
Collapse
Affiliation(s)
- Samira Rasaneh
- Department of Medical Physics, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14115-331, Tehran, Iran
| | | | | | | |
Collapse
|
23
|
Radiolabeling of trastuzumab with 177Lu via DOTA, a new radiopharmaceutical for radioimmunotherapy of breast cancer. Nucl Med Biol 2009; 36:363-9. [PMID: 19423003 DOI: 10.1016/j.nucmedbio.2009.01.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Revised: 01/18/2009] [Accepted: 01/31/2009] [Indexed: 11/20/2022]
Abstract
AIM Trastuzumab is a monoclonal antibody that is used in treating breast cancer. We labeled this monoclonal antibody with lutetium-177 and performed in vitro quality control tests as a first step in the production of a new radiopharmaceutical. MATERIAL AND METHODS Trastuzumab was labeled with lutetium-177 using DOTA as chelator. Radiochemical purity and stability in buffer and human blood serum were determined using thin layer chromatography. Immunoreactivity and toxicity of the complex were tested on MCF7 breast cancer cell line. RESULTS The radiochemical purity of the complex was 96+/-0.9%. The stabilities in phosphate buffer and in human blood serum at 96 h postpreparation were 93+/-1.2% and 85+/-3.5%, respectively. The immunoreactivity of the complex was 89+/-1.4%. At a concentration of 1 nM, the complex killed 70+/-3% of MCF7 cells. At 1.9 nM, 90+/-5% of the cells were killed. CONCLUSIONS The results showed that the new complex could be considered for further evaluation in animals and possibly in humans as a new radiopharmaceutical for use in radioimmunotherapy against breast cancer.
Collapse
|
24
|
Hens M, Vaidyanathan G, Welsh P, Zalutsky MR. Labeling internalizing anti-epidermal growth factor receptor variant III monoclonal antibody with (177)Lu: in vitro comparison of acyclic and macrocyclic ligands. Nucl Med Biol 2009; 36:117-28. [PMID: 19217523 PMCID: PMC2663407 DOI: 10.1016/j.nucmedbio.2008.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2008] [Revised: 10/29/2008] [Accepted: 11/01/2008] [Indexed: 12/01/2022]
Abstract
INTRODUCTION The monoclonal antibody (mAb) L8A4, reactive with the epidermal growth factor receptor variant III (EGFRvIII), internalizes rapidly in glioma cells after receptor binding. Combining this tumor-specific mAb with the low-energy beta-emitter (177)Lu would be an attractive approach for brain tumor radioimmunotherapy, provided that trapping of the radionuclide in tumor cells after mAb intracellular processing could be maximized. MATERIALS AND METHODS L8A4 mAb was labeled with (177)Lu using the acyclic ligands [(R)-2-amino-3-(4-isothiocyanatophenyl)propyl]-trans-(S,S)-cyclohexane-1,2-diamine-pentaacetic acid (CHX-A''-DTPA), 2-(4-isothiocyanatobenzyl)-diethylenetriaminepenta-acetic acid (pSCN-Bz-DTPA) and 2-(4-isothiocyanatobenzyl)-6-methyldiethylenetriaminepentaacetic acid (1B4M-DTPA), and the macrocyclic ligands S-2-(4-isothiocyanatobenzyl)-1,4,7,10-tetraazacyclododecane-tetraacetic acid (C-DOTA) and alpha-(5-isothiocyanato-2-methoxyphenyl)-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (MeO-DOTA). Paired-label internalization and cellular processing assays were performed on EGFRvIII-expressing U87.DeltaEGFR glioma cells over 24 h to directly compare (177)Lu-labeled L8A4 to L8A4 labeled with (125)I using either iodogen or N-succinimidyl 4-guanidinomethyl-3-[(125)I]iodobenzoate ([(125)I]SGMIB). In order to facilitate comparison of labeling methods, the primary parameter evaluated was the ratio of (177)Lu to (125)I activity retained in U87.DeltaEGFR cells. RESULTS All chelates demonstrated higher retention of internalized activity compared with mAb labeled using iodogen, with (177)Lu/(125)I ratios of >20 observed for the three DTPA chelates at 24 h. When compared to L8A4 labeled using SGMIB, except for MeO-DOTA, internalized activity for (125)I was higher than (177)Lu from 1-8 h with the opposite behavior observed thereafter. At 24 h, (177)Lu/(125)I ratios were between 1.5 and 3, with higher values observed for the three DTPA chelates. CONCLUSIONS The nature of the chelate used to label this internalizing mAb with (177)Lu influenced intracellular retention in vitro, although at early time points, only MeO-DOTA provided more favorable results than radioiodination of the mAb via SGMIB.
Collapse
Affiliation(s)
- Marc Hens
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | | | - Phil Welsh
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Michael R. Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
25
|
Elmore CS. Chapter 25 The Use of Isotopically Labeled Compounds in Drug Discovery. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2009. [DOI: 10.1016/s0065-7743(09)04425-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
26
|
Rich RL, Myszka DG. Survey of the year 2007 commercial optical biosensor literature. J Mol Recognit 2008; 21:355-400. [DOI: 10.1002/jmr.928] [Citation(s) in RCA: 144] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
27
|
Lindegren S, Frost S, Bäck T, Haglund E, Elgqvist J, Jensen H. Direct procedure for the production of 211At-labeled antibodies with an epsilon-lysyl-3-(trimethylstannyl)benzamide immunoconjugate. J Nucl Med 2008; 49:1537-45. [PMID: 18703591 DOI: 10.2967/jnumed.107.049833] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED (211)At-labeled tumor-specific antibodies have long been considered for the treatment of disseminated cancer. However, the limited availability of the nuclide and the poor efficacy of labeling procedures at clinical activity levels present major obstacles to their use. This study evaluated a procedure for the direct astatination of antibodies for the production of clinical activity levels. METHODS The monoclonal antibody trastuzumab was conjugated with the reagent N-succinimidyl-3-(trimethylstannyl)benzoate, and the immunoconjugate was labeled with astatine. Before astatination of the conjugated antibody, the nuclide was activated with N-iodosuccinimide. The labeling reaction was evaluated in terms of reaction time, volume of reaction solvent, immunoconjugate concentration, and applied activity. The quality of the astatinated antibodies was determined by in vitro analysis and biodistribution studies in nude mice. RESULTS The reaction proceeded almost instantaneously, and the results indicated a low dependence on immunoconjugate concentration and applied activity. Radiochemical labeling yields were in the range of 68%-81%, and a specific radioactivity of up to 1 GBq/mg could be achieved. Stability and radiochemical purity were equal to or better than those attained with a conventional 2-step procedure. Dissociation constants for directly astatinated, conventionally astatinated, and radioiodinated trastuzumab were 1.0+/-0.06 (mean+/-SD), 0.44+/-0.06, and 0.29+/-0.02 nM, respectively. The tissue distribution in non-tumor-bearing nude mice revealed only minor differences in organ uptake relative to that obtained with the conventional method. CONCLUSION The direct astatination procedure enables the high-yield production of astatinated antibodies with radioactivity in the amounts required for clinical applications.
Collapse
Affiliation(s)
- Sture Lindegren
- Department of Radiation Physics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | | | | | | | | | | |
Collapse
|