1
|
El-Kadiry AEH, Beaudoin S, Plouffe S, Rafei M. Accum™ Technology: A Novel Conjugable Primer for Onco-Immunotherapy. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123807. [PMID: 35744930 PMCID: PMC9227040 DOI: 10.3390/molecules27123807] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/10/2022] [Accepted: 06/11/2022] [Indexed: 11/20/2022]
Abstract
Compromised activity is a common impediment for biologics requiring endosome trafficking into target cells. In cancer cells, antibody-drug conjugates (ADCs) are trapped in endosomes or subsequently pumped extracellularly, leading to a reduction in intracellular accumulation. In subsets of dendritic cells (DCs), endosome-engulfed antigens face non-specific proteolysis and collateral damage to epitope immunogenicity before proteasomal processing and subsequent surface presentation. To bypass these shortcomings, we devised Accum™, a conjugable biotechnology harboring cholic acid (ChAc) and a nuclear localization signal (NLS) sequence for endosome escape and prompt nuclear targeting. Combined, these mechanisms culminate in enhanced intracellular accumulation and functionalization of coupled biologics. As proof-of-principle, we have biochemically characterized Accum, demonstrating its adaptability to ADCs or antigens in different cancer settings. Additionally, we have validated that endosome escape and nuclear routing are indispensable for effective intracellular accumulation and guaranteed target cell selectivity. Importantly, we have demonstrated that the unique mechanism of action of Accum translates into enhanced tumor cytotoxicity when coupled to ADCs, and durable therapeutic and prophylactic anti-cancer immunogenicity when coupled to tumor antigens. As more pre-clinical evidence accumulates, the adaptability, unique mechanism of action, and high therapeutic potency of Accum signal a promising transition into clinical investigations in the context of onco-immunotherapy.
Collapse
Affiliation(s)
- Abed El-Hakim El-Kadiry
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC H3T 1J4, Canada;
| | - Simon Beaudoin
- Defence Therapeutics Inc., Research and Development Branch, Vancouver, BC V6C 3L6, Canada; (S.B.); (S.P.)
| | - Sebastien Plouffe
- Defence Therapeutics Inc., Research and Development Branch, Vancouver, BC V6C 3L6, Canada; (S.B.); (S.P.)
| | - Moutih Rafei
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC H3T 1J4, Canada;
- Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Molecular Biology Program, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Correspondence: ; Tel.: +1-(514)-343-6931
| |
Collapse
|
2
|
Sauter M, Strieker M, Kleist C, Wischnjow A, Daniel V, Altmann A, Haberkorn U, Mier W. Improving antibody-based therapies by chemical engineering of antibodies with multimeric cell-penetrating peptides for elevated intracellular delivery. J Control Release 2020; 322:200-208. [PMID: 32184098 DOI: 10.1016/j.jconrel.2020.03.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 12/22/2022]
Abstract
Monoclonal antibodies (mAbs) are increasingly exploited as vehicles for the targeted delivery of cytotoxic drugs. In antibody-drug conjugates (ADCs) antibodies specifically deliver cytotoxic compounds to cancer cells. Here, we present a technology for elevating the intracellular delivery of antibodies by the conjugation of tetrameric cell-penetrating peptides (tCPPs). The solid phase synthesis of tCPPs and their application in a chemical modification strategy for mAbs provides constructs that attain up to fourfold elevated internalization rates while retaining the mAbs target specificity. The antigen independent internalization is accompanied by beneficial pharmacokinetics limiting off-target accumulation. Applicability was proven for matuzumab, trastuzumab and the ADC Kadcyla®. Cytotoxicity studies of tCPP-conjugates of Kadcyla® resulted in a sixfold increased cytotoxicity proving the potential of chemical modification strategies to extend the applicability of biologicals. This constitutes a significant step towards next-generation antibody-based therapeutics.
Collapse
Affiliation(s)
- Max Sauter
- Department of Nuclear Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Matthias Strieker
- Department of Nuclear Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Christian Kleist
- Department of Nuclear Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Artjom Wischnjow
- Department of Nuclear Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Volker Daniel
- Institute of Immunology, Heidelberg University Hospital, Im Neuenheimer Feld 305, 69120 Heidelberg, Germany
| | - Annette Altmann
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 350, 69120 Heidelberg, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 350, 69120 Heidelberg, Germany
| | - Walter Mier
- Department of Nuclear Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany.
| |
Collapse
|
3
|
Peltek OO, Muslimov AR, Zyuzin MV, Timin AS. Current outlook on radionuclide delivery systems: from design consideration to translation into clinics. J Nanobiotechnology 2019; 17:90. [PMID: 31434562 PMCID: PMC6704557 DOI: 10.1186/s12951-019-0524-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/14/2019] [Indexed: 02/06/2023] Open
Abstract
Radiopharmaceuticals have proven to be effective agents, since they can be successfully applied for both diagnostics and therapy. Effective application of relevant radionuclides in pre-clinical and clinical studies depends on the choice of a sufficient delivery platform. Herein, we provide a comprehensive review on the most relevant aspects in radionuclide delivery using the most employed carrier systems, including, (i) monoclonal antibodies and their fragments, (ii) organic and (iii) inorganic nanoparticles, and (iv) microspheres. This review offers an extensive analysis of radionuclide delivery systems, the approaches of their modification and radiolabeling strategies with the further prospects of their implementation in multimodal imaging and disease curing. Finally, the comparative outlook on the carriers and radionuclide choice, as well as on the targeting efficiency of the developed systems is discussed.
Collapse
Affiliation(s)
- Oleksii O Peltek
- Russian Research Center of Radiology and Surgical Technologies (RRCRST) of Ministry of Public Health, Leningradskaya Street 70 Pesochny, Saint-Petersburg, 197758, Russian Federation
| | - Albert R Muslimov
- Russian Research Center of Radiology and Surgical Technologies (RRCRST) of Ministry of Public Health, Leningradskaya Street 70 Pesochny, Saint-Petersburg, 197758, Russian Federation
| | - Mikhail V Zyuzin
- Faculty of Physics and Engineering, ITMO University, St. Petersburg, 197101, Russia
| | - Alexander S Timin
- Russian Research Center of Radiology and Surgical Technologies (RRCRST) of Ministry of Public Health, Leningradskaya Street 70 Pesochny, Saint-Petersburg, 197758, Russian Federation.
- Research School of Chemical and Biomedical Engineering, National Research Tomsk Polytechnic University, Lenin Avenue 30, Tomsk, 634050, Russia.
| |
Collapse
|
4
|
Singh K, Ejaz W, Dutta K, Thayumanavan S. Antibody Delivery for Intracellular Targets: Emergent Therapeutic Potential. Bioconjug Chem 2019; 30:1028-1041. [PMID: 30830750 PMCID: PMC6470022 DOI: 10.1021/acs.bioconjchem.9b00025] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Proteins have sparked fast growing interest as biological therapeutic agents for several diseases. Antibodies, in particular, carry an enormous potential as drugs owing to their remarkable target specificity and low immunogenicity. Although the market has numerous antibodies directed toward extracellular targets, their use in targeting therapeutically important intracellular targets is limited by their inability to cross cellular membrane. Realizing the potential for antibody therapy in disease treatment, progress has been made in the development of methods to deliver antibodies intracellularly. In this review, we address various platforms for delivery of antibodies and their merits and drawbacks.
Collapse
|
5
|
Davydova M, Dewaele Le Roi G, Adumeau P, Zeglis BM. Synthesis and Bioconjugation of Thiol-Reactive Reagents for the Creation of Site-Selectively Modified Immunoconjugates. J Vis Exp 2019. [PMID: 30907883 DOI: 10.3791/59063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Maleimide-bearing bifunctional probes have been employed for decades for the site-selective modification of thiols in biomolecules, especially antibodies. Yet maleimide-based conjugates display limited stability in vivo because the succinimidyl thioether linkage can undergo a retro-Michael reaction. This, of course, can lead to the release of the radioactive payload or its exchange with thiol-bearing biomolecules in circulation. Both of these processes can produce elevated activity concentrations in healthy organs as well as decreased activity concentrations in target tissues, resulting in reduced imaging contrast and lower therapeutic ratios. In 2018, we reported the creation of a modular, stable, and easily accessible phenyloxadiazolyl methyl sulfone reagent - dubbed 'PODS' - as a platform for thiol-based bioconjugations. We have clearly demonstrated that PODS-based site-selective bioconjugations reproducibly and robustly create homogenous, well-defined, highly immunoreactive, and highly stable radioimmunoconjugates. Furthermore, preclinical experiments in murine models of colorectal cancer have shown that these site-selectively labeled radioimmunoconjugates exhibit far superior in vivo performance compared to radiolabeled antibodies synthesized via maleimide-based conjugations. In this protocol, we will describe the four-step synthesis of PODS, the creation of a bifunctional PODS-bearing variant of the ubiquitous chelator DOTA (PODS-DOTA), and the conjugation of PODS-DOTA to the HER2-targeting antibody trastuzumab.
Collapse
Affiliation(s)
- Maria Davydova
- Department of Chemistry, Hunter College of the City University of New York
| | - Guillaume Dewaele Le Roi
- Department of Chemistry, Hunter College of the City University of New York; Ph.D. Program in Chemistry, Graduate Center of the City University of New York
| | - Pierre Adumeau
- Department of Chemistry, Hunter College of the City University of New York
| | - Brian M Zeglis
- Department of Chemistry, Hunter College of the City University of New York; Ph.D. Program in Chemistry, Graduate Center of the City University of New York; Department of Radiology, Memorial Sloan Kettering Cancer Center; Department of Radiology, Weill Cornell Medical College;
| |
Collapse
|
6
|
Morais M, Ma MT. Site-specific chelator-antibody conjugation for PET and SPECT imaging with radiometals. DRUG DISCOVERY TODAY. TECHNOLOGIES 2018; 30:91-104. [PMID: 30553525 PMCID: PMC6291455 DOI: 10.1016/j.ddtec.2018.10.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 11/17/2022]
Abstract
Antibodies and their derivatives radiolabelled with positron- and gamma-emitting radiometals enable sensitive and quantitative molecular Positron Emission Tomography (PET) and Single Photon Emission Computed Tomography (SPECT) imaging of antibody distribution in vivo. Chelators that are covalently attached to antibodies allow radiolabelling with metallic PET and SPECT radioisotopes. Conventional strategies for chelator-protein conjugation generate heterogeneous mixtures of bioconjugates that can exhibit reduced affinity for their receptor targets, and undesirable biodistribution and pharmacokinetics. Recent advances in bioconjugation technology enable site-specific modification to generate well-defined constructs with superior properties. Herein we survey existing site-specific chelator-protein conjugation methods. These include chelator attachment to cysteines/disulfide bonds or the glycan region of the antibody, enzyme-mediated chelator conjugation, and incorporation of sequences of amino acids that chelate the radiometal. Such technology will allow better use of PET and SPECT imaging in the development of antibody-based therapies.
Collapse
Affiliation(s)
- Mauricio Morais
- School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, United Kingdom.
| | - Michelle T Ma
- School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| |
Collapse
|
7
|
Beaudoin S, Paquette M, Fafard-Couture L, Tremblay MA, Lecomte R, Guérin B, Leyton JV. Initial Evaluation of Antibody-conjugates Modified with Viral-derived Peptides for Increasing Cellular Accumulation and Improving Tumor Targeting. J Vis Exp 2018. [PMID: 29578523 DOI: 10.3791/55440] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Antibody-conjugates (ACs) modified with virus-derived peptides are a potentially powerful class of tumor cell delivery agents for molecular payloads used in cancer treatment and imaging due to increased cellular accumulation over current ACs. During early AC in vitro development, fluorescence techniques and radioimmunoassays are sufficient for determining intracellular localization, accumulation efficiency, and target cell specificity. Currently, there is no consensus on standardized methods for preparing cells for evaluating AC intracellular accumulation and localization. The initial testing of ACs modified with virus-derived peptides is critical especially if several candidates have been constructed. Determining intracellular accumulation by fluorescence can be affected by background signal from ACs at the cell surface and complicate the interpretation of accumulation. For radioimmunoassays, typically treated cells are fractionated and the radioactivity in different cell compartments measured. However, cell lysis varies from cell to cell and often nuclear and cytoplasmic compartments are not adequately isolated. This can produce misleading data on payload delivery properties. The intravenous injection of radiolabeled virus-derived peptide-modified ACs in tumor bearing mice followed by radionuclide imaging is a powerful method for determining tumor targeting and payload delivery properties at the in vivo phase of development. However, this is a relatively recent advancement and few groups have evaluated virus-derived peptide-modified ACs in this manner. We describe the processing of treated cells to more accurately evaluate virus-derived peptide-modified AC accumulation when using confocal microscopy and radioimmunoassays. Specifically, a method for trypsinizing cells to remove cell surface bound ACs. We also provide a method for improving cellular fractionation. Lastly, this protocol provides an in vivo method using positron emission tomography (PET) for evaluating initial tumor targeting properties in tumor-bearing mice. We use the radioisotope 64Cu (t1/2 = 12.7 h) as an example payload in this protocol.
Collapse
Affiliation(s)
- Simon Beaudoin
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke
| | - Michel Paquette
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke
| | | | - Mylene A Tremblay
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke
| | - Roger Lecomte
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke; Sherbrooke Molecular Imaging Center (CIMS), Université de Sherbrooke; Sherbrooke Institute of Pharmacology
| | - Brigitte Guérin
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke; Sherbrooke Molecular Imaging Center (CIMS), Université de Sherbrooke; Sherbrooke Institute of Pharmacology
| | - Jeffrey V Leyton
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke; Sherbrooke Molecular Imaging Center (CIMS), Université de Sherbrooke; Sherbrooke Institute of Pharmacology;
| |
Collapse
|
8
|
Paquette M, Beaudoin S, Tremblay MA, Jean S, Lopez AF, Lecomte R, Guérin B, Bentourkia M, Sabbagh R, Leyton JV. NLS-Cholic Acid Conjugation to IL-5Rα-Specific Antibody Improves Cellular Accumulation and In Vivo Tumor-Targeting Properties in a Bladder Cancer Model. Bioconjug Chem 2018; 29:1352-1363. [PMID: 29433309 DOI: 10.1021/acs.bioconjchem.8b00077] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Receptor-mediated internalization followed by trafficking and degradation of antibody-conjugates (ACs) via the endosomal-lysosomal pathway is the major mechanism for delivering molecular payloads inside target tumor cells. Although a mainstay for delivering payloads with clinically approved ACs in cancer treatment and imaging, tumor cells are often able to decrease intracellular payload concentrations and thereby reduce the effectiveness of the desired application. Thus, increasing payload intracellular accumulation has become a focus of attention for designing next-generation ACs. We developed a composite compound (ChAcNLS) that enables ACs to escape endosome entrapment and route to the nucleus resulting in the increased intracellular accumulation as an interleukin-5 receptor α-subunit (IL-5Rα)-targeted agent for muscle invasive bladder cancer (MIBC). We constructed 64Cu-A14-ChAcNLS, 64Cu-A14-NLS, and 64Cu-A14 and evaluated their performance by employing mechanistic studies for endosome escape coupled to nuclear routing and determining whether this delivery system results in improved 64Cu cellular accumulation. ACs consisting of ∼20 ChAcNLS or NLS moieties per 64Cu-A14 were prepared in good yield, high monomer content, and maintaining high affinity for IL-5Rα. Confocal microscopy analysis demonstrated ChAcNLS mediated efficient endosome escape and nuclear localization. 64Cu-A14-ChAcNLS increased 64Cu cellular accumulation in HT-1376 and HT-B9 cells relative to 64Cu-A14 and 64Cu-A14-NLS. In addition, we tested 64Cu-A14-ChAcNLS in vivo to evaluate its tissue distribution properties and, ultimately, tumor uptake and targeting. A model of human IL-5Rα MIBC was developed by implanting NOD/SCID mice with subcutaneous HT-1376 or HT-B9MIBC tumors, which grow containing high and low IL-5Rα-positive tumor cell densities, respectively. ACs were intravenously injected, and daily blood sampling, biodistribution at 48 and 96 h, and positron emission tomography (PET) at 24 and 48 h were performed. Region of interest (ROI) analysis was also performed on reconstructed PET images. Pharmacokinetic analysis and biodistribution studies showed that 64Cu-A14-ChAcNLS had faster clearance rates from the blood and healthy organs relative to 64Cu-A14. However, 64Cu-A14-ChAcNLS maintained comparable tumor accumulation relative to 64Cu-A14. This resulted in 64Cu-A14-ChAcNLS having superior tumor/normal tissue ratios at both 48 and 96 h biodistribution time points. Visualization of AC distribution by PET and ROI analysis confirmed that 64Cu-A14-ChAcNLS had improved targeting of MIBC tumor relative to 64Cu-A14. In addition, 64Cu-A14 modified with only NLS had poor tumor targeting. This was a result of poor tumor uptake due to extremely rapid clearance. Thus, the overall findings in this model of human IL-5Rα-positive MIBC describe an endosome escape-nuclear localization cholic-acid-linked peptide that substantially enhances AC cellular accumulation and tumor targeting.
Collapse
Affiliation(s)
| | | | | | | | - Angel F Lopez
- The Centre for Cancer Biology, SA Pathology , The University of South Australia , Frome Road , Adelaide , South Australia 5000 , Australia
| | - Roger Lecomte
- Sherbrooke Molecular Imaging Centre (CIMS), Centre de Recherche du CHUS , UdeS , 3001 12 Avenue Nord , Sherbrooke , Québec J1H 5N4 , Canada.,Sherbrooke Pharmacology Institute , 3001 12 Avenue Nord , Sherbrooke , Québec J1H 5N4 , Canada
| | - Brigitte Guérin
- Sherbrooke Molecular Imaging Centre (CIMS), Centre de Recherche du CHUS , UdeS , 3001 12 Avenue Nord , Sherbrooke , Québec J1H 5N4 , Canada.,Sherbrooke Pharmacology Institute , 3001 12 Avenue Nord , Sherbrooke , Québec J1H 5N4 , Canada
| | - M'hamed Bentourkia
- Sherbrooke Molecular Imaging Centre (CIMS), Centre de Recherche du CHUS , UdeS , 3001 12 Avenue Nord , Sherbrooke , Québec J1H 5N4 , Canada.,Sherbrooke Pharmacology Institute , 3001 12 Avenue Nord , Sherbrooke , Québec J1H 5N4 , Canada
| | | | - Jeffrey V Leyton
- Sherbrooke Molecular Imaging Centre (CIMS), Centre de Recherche du CHUS , UdeS , 3001 12 Avenue Nord , Sherbrooke , Québec J1H 5N4 , Canada.,Sherbrooke Pharmacology Institute , 3001 12 Avenue Nord , Sherbrooke , Québec J1H 5N4 , Canada
| |
Collapse
|
9
|
Okuda-Shinagawa NM, Moskalenko YE, Junqueira HC, Baptista M, Marques CM, Machini MT. Fluorescent and Photosensitizing Conjugates of Cell-Penetrating Peptide TAT(47-57): Design, Microwave-Assisted Synthesis at 60 °C, and Properties. ACS OMEGA 2017; 2:8156-8166. [PMID: 30023576 PMCID: PMC6044864 DOI: 10.1021/acsomega.7b01127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/30/2017] [Indexed: 05/04/2023]
Abstract
Conjugates based on cell-penetrating peptides (CPPs) are scientifically relevant owing to their structural complexity; their ability to enter cells and deliver drugs, labels, antioxidants, bioactive compounds, or DNA fragments; and, consequently, their potential for application in research and biomedicine. In this study, carboxyamidated fluorescently labeled conjugates FAM-GG-TAT(47-57)-NH2 and FAM-PEG6-TAT(47-57)-NH2 and photosensitizer-labeled conjugate Chk-PEG6-TAT(47-57)-NH2 [where TAT(47-57) is the CPP, 5(6)-carboxyfluorescein is the (FAM) fluorophore, chlorin k (Chk) is the photosensitizer, and the dipeptide glycyl-glycine (GG) or hexaethylene glycol (PEG6) is the spacer] were originally designed, prepared, and fully characterized. Practically, all chemical reactions of the synthetic steps (peptide synthesis, spacer incorporation, and conjugation) were microwave-assisted at 60 °C using optimized protocols to give satisfying yields and high-quality products. Detailed analyses of the conjugates using spectrofluorimetry and singlet oxygen detection showed that they display photophysical properties typical of FAM or Chk. Anticandidal activity assays showed that not only this basic property of TAT(47-57) was preserved in the conjugates but also that the minimal inhibitory concentration was slightly reduced for cells incubated with PS-bearing conjugate Chk-PEG6-TAT(47-57)-NH2. Overall, these results indicated that the synthetic approach on-resin assisted by microwaves at 60 °C is simple, straightforward, selective, metal-free, sufficiently fast, cleaner, and more cost-effective than those previously used for preparing this type of macromolecule. Furthermore, such new data show that microwaves at 60 °C and/or conjugation did not harm the integrity of the conjugates' constituents. Therefore, FAM-GG-TAT(47-57)-NH2, FAM-PEG6-TAT(47-57)-NH2, and Chk-PEG6-TAT(47-57)-NH2 have high potential for practical applications in biochemistry, biophysics, and therapeutics.
Collapse
Affiliation(s)
- Nancy M. Okuda-Shinagawa
- Department
of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes, 748, Cidade Universitária, Butantã, 05508-000 São
Paulo, SP, Brazil
| | - Yulia E. Moskalenko
- Department
of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes, 748, Cidade Universitária, Butantã, 05508-000 São
Paulo, SP, Brazil
| | - Helena C. Junqueira
- Department
of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes, 748, Cidade Universitária, Butantã, 05508-000 São
Paulo, SP, Brazil
| | - Maurício
S. Baptista
- Department
of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes, 748, Cidade Universitária, Butantã, 05508-000 São
Paulo, SP, Brazil
| | - Carlos M. Marques
- Department
of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes, 748, Cidade Universitária, Butantã, 05508-000 São
Paulo, SP, Brazil
- Institut
Charles Sadron, Université de Strasbourg,
UPR22-CNRS, 23, rue du
Loess, BP 84047, 67034 Strasbourg Cedex 2, Strasbourg, France
| | - M. Terêsa Machini
- Department
of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes, 748, Cidade Universitária, Butantã, 05508-000 São
Paulo, SP, Brazil
| |
Collapse
|
10
|
Adumeau P, Sharma SK, Brent C, Zeglis BM. Site-Specifically Labeled Immunoconjugates for Molecular Imaging--Part 1: Cysteine Residues and Glycans. Mol Imaging Biol 2016; 18:1-17. [PMID: 26754790 PMCID: PMC4722084 DOI: 10.1007/s11307-015-0919-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Due to their remarkable selectivity and specificity for cancer biomarkers, immunoconjugates have emerged as extremely promising vectors for the delivery of diagnostic radioisotopes and fluorophores to malignant tissues. Paradoxically, however, these tools for precision medicine are synthesized in a remarkably imprecise way. Indeed, the vast majority of immunoconjugates are created via the random conjugation of bifunctional probes (e.g., DOTA-NCS) to amino acids within the antibody (e.g., lysines). Yet antibodies have multiple copies of these residues throughout their macromolecular structure, making control over the location of the conjugation reaction impossible. This lack of site specificity can lead to the formation of poorly defined, heterogeneous immunoconjugates with suboptimal in vivo behavior. Over the past decade, interest in the synthesis and development of site-specifically labeled immunoconjugates—both antibody-drug conjugates as well as constructs for in vivo imaging—has increased dramatically, and a number of reports have suggested that these better defined, more homogeneous constructs exhibit improved performance in vivo compared to their randomly modified cousins. In this two-part review, we seek to provide an overview of the various methods that have been developed to create site-specifically modified immunoconjugates for positron emission tomography, single photon emission computed tomography, and fluorescence imaging. We will begin with an introduction to the structure of antibodies and antibody fragments. This is followed by the core of the work: sections detailing the four different approaches to site-specific modification strategies based on cysteine residues, glycans, peptide tags, and unnatural amino acids. These discussions will be divided into two installments: cysteine residues and glycans will be detailed in Part 1 of the review, while peptide tags and unnatural amino acids will be addressed in Part 2. Ultimately, we sincerely hope that this review fosters interest and enthusiasm for site-specific immunoconjugates within the nuclear medicine and molecular imaging communities.
Collapse
Affiliation(s)
- Pierre Adumeau
- Department of Chemistry and Biochemistry, Hunter College and the Graduate Center of the City University of New York, 413 East 69th Street, New York, NY, 10021, USA
| | - Sai Kiran Sharma
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY10065, NY, USA
| | - Colleen Brent
- Department of Chemistry and Biochemistry, Hunter College and the Graduate Center of the City University of New York, 413 East 69th Street, New York, NY, 10021, USA
| | - Brian M Zeglis
- Department of Chemistry and Biochemistry, Hunter College and the Graduate Center of the City University of New York, 413 East 69th Street, New York, NY, 10021, USA.
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY10065, NY, USA.
| |
Collapse
|
11
|
Cai Z, Kwon YL, Reilly RM. Monte Carlo N-Particle (MCNP) Modeling of the Cellular Dosimetry of 64Cu: Comparison with MIRDcell S Values and Implications for Studies of Its Cytotoxic Effects. J Nucl Med 2016; 58:339-345. [PMID: 27660146 DOI: 10.2967/jnumed.116.175695] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 09/01/2016] [Indexed: 12/18/2022] Open
Abstract
64Cu emits positrons as well as β- particles and Auger and internal conversion electrons useful for radiotherapy. Our objective was to model the cellular dosimetry of 64Cu under different geometries commonly used to study the cytotoxic effects of 64Cu. METHODS Monte Carlo N-Particle (MCNP) was used to simulate the transport of all particles emitted by 64Cu from the cell surface (CS), cytoplasm (Cy), or nucleus (N) of a single cell; monolayer in a well (radius = 0.32-1.74 cm); or a sphere (radius = 50-6,000 μm) of cells to calculate S values. The radius of the cell and N ranged from 5 to 12 μm and 2 to 11 μm, respectively. S values were obtained by MIRDcell for comparison. MCF7/HER2-18 cells were exposed in vitro to 64Cu-labeled trastuzumab. The subcellular distribution of 64Cu was measured by cell fractionation. The surviving fraction was determined in a clonogenic assay. RESULTS The relative differences of MCNP versus MIRDcell self-dose S values (Sself) for 64Cu ranged from -0.2% to 3.6% for N to N (SN←N), 2.3% to 8.6% for Cy to N (SN←Cy), and -12.0% to 7.3% for CS to N (SN←CS). The relative differences of MCNP versus MIRDcell cross-dose S values were 25.8%-30.6% for a monolayer and 30%-34% for a sphere, respectively. The ratios of SN←N versus SN←Cy and SN←Cy versus SN←CS decreased with increasing ratio of the N of the cell versus radius of the cell and the size of the monolayer or sphere. The surviving fraction of MCF7 /: HER2-18 cells treated with 64Cu-labeled trastuzumab (0.016-0.368 MBq/μg, 67 nM) for 18 h versus the absorbed dose followed a linear survival curve with α = 0.51 ± 0.05 Gy-1 and R2 = 0.8838. This is significantly different from the linear quadratic survival curve of MCF7 /: HER2-18 cells exposed to γ-rays. CONCLUSION MCNP- and MIRDcell-calculated S values agreed well. 64Cu in the N increases the dose to the N in isolated single cells but has less effect in a cell monolayer or small cluster of cells simulating a micrometastasis, and little effect in a sphere analogous to a tumor xenograft compared with 64Cu in the Cy or on the CS. The dose deposited by 64Cu is less effective for cell killing than γ-rays.
Collapse
Affiliation(s)
- Zhongli Cai
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Yongkyu Luke Kwon
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Raymond M Reilly
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada .,Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada.,Toronto General Research Institute, Toronto, Ontario, Canada; and.,Joint Department of Medical Imaging, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
12
|
Beaudoin S, Rondeau A, Martel O, Bonin MA, van Lier JE, Leyton JV. ChAcNLS, a Novel Modification to Antibody-Conjugates Permitting Target Cell-Specific Endosomal Escape, Localization to the Nucleus, and Enhanced Total Intracellular Accumulation. Mol Pharm 2016; 13:1915-26. [PMID: 27112376 DOI: 10.1021/acs.molpharmaceut.6b00075] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The design of antibody-conjugates (ACs) for delivering molecules for targeted applications in humans has sufficiently progressed to demonstrate clinical efficacy in certain malignancies and reduced systemic toxicity that occurs with standard nontargeted therapies. One area that can advance clinical success for ACs will be to increase their intracellular accumulation. However, entrapment and degradation in the endosomal-lysosomal pathway, on which ACs are reliant for the depositing of their molecular payload inside target cells, leads to reduced intracellular accumulation. Innovative approaches that can manipulate this pathway may provide a strategy for increasing accumulation. We hypothesized that escape from entrapment inside the endosomal-lysosomal pathway and redirected trafficking to the nucleus could be an effective approach to increase intracellular AC accumulation in target cells. Cholic acid (ChAc) was coupled to the peptide CGYGPKKKRKVGG containing the nuclear localization sequence (NLS) from SV-40 large T-antigen, which is termed ChAcNLS. ChAcNLS was conjugated to the mAb 7G3 (7G3-ChAcNLS), which has nanomolar affinity for the cell-surface leukemic antigen interleukin-3 receptor-α (IL-3Rα). Our aim was to determine whether 7G3-ChAcNLS increased intracellular accumulation while retaining nanomolar affinity and IL-3Rα-positive cell selectivity. Competition ELISA and cell treatment assays were performed. Cell fractionation, confocal microscopy, flow cytometry, and Western blot techniques were used to determine the level of antibody accumulation inside cells and in corresponding nuclei. In addition, the radioisotope copper-64 ((64)Cu) was also utilized as a surrogate molecular cargo to evaluate nuclear and intracellular accumulation by radioactivity counting. 7G3-ChAcNLS effectively escaped endosome entrapment and degradation resulting in a unique intracellular distribution pattern. mAb modification with ChAcNLS maintained 7G3 nM affinity and produced high selectivity for IL-3Rα-positive cells. In contrast, 7G3 ACs with the ability to either escape endosome entrapment or traffic to the nucleus was not superior to 7G3-ChAcNLS for increasing intracellular accumulation. Transportation of (64)Cu when complexed to 7G3-ChAcNLS also resulted in increased nuclear and intracellular radioactivity accumulation. Thus, ChAcNLS is a novel mAb functionalizing technology that demonstrates its ability to increase AC intracellular accumulation in target cells through escaping endosome entrapment coupled to nuclear trafficking.
Collapse
Affiliation(s)
- Simon Beaudoin
- Departément de médecine nucléaire et radiobiologie, ‡Plateforme de synthèse de peptides et de sondes d'imageries, Faculté de médecine et sciences de la santé, and §Centre d'imagerie moléculaire de Sherbrooke (CIMS), Université de Sherbrooke , 3001 12e Avenue Nord, Sherbrooke, Québec J1H5N4, Canada
| | - Andreanne Rondeau
- Departément de médecine nucléaire et radiobiologie, ‡Plateforme de synthèse de peptides et de sondes d'imageries, Faculté de médecine et sciences de la santé, and §Centre d'imagerie moléculaire de Sherbrooke (CIMS), Université de Sherbrooke , 3001 12e Avenue Nord, Sherbrooke, Québec J1H5N4, Canada
| | - Olivier Martel
- Departément de médecine nucléaire et radiobiologie, ‡Plateforme de synthèse de peptides et de sondes d'imageries, Faculté de médecine et sciences de la santé, and §Centre d'imagerie moléculaire de Sherbrooke (CIMS), Université de Sherbrooke , 3001 12e Avenue Nord, Sherbrooke, Québec J1H5N4, Canada
| | - Marc-Andre Bonin
- Departément de médecine nucléaire et radiobiologie, ‡Plateforme de synthèse de peptides et de sondes d'imageries, Faculté de médecine et sciences de la santé, and §Centre d'imagerie moléculaire de Sherbrooke (CIMS), Université de Sherbrooke , 3001 12e Avenue Nord, Sherbrooke, Québec J1H5N4, Canada
| | - Johan E van Lier
- Departément de médecine nucléaire et radiobiologie, ‡Plateforme de synthèse de peptides et de sondes d'imageries, Faculté de médecine et sciences de la santé, and §Centre d'imagerie moléculaire de Sherbrooke (CIMS), Université de Sherbrooke , 3001 12e Avenue Nord, Sherbrooke, Québec J1H5N4, Canada
| | - Jeffrey V Leyton
- Departément de médecine nucléaire et radiobiologie, ‡Plateforme de synthèse de peptides et de sondes d'imageries, Faculté de médecine et sciences de la santé, and §Centre d'imagerie moléculaire de Sherbrooke (CIMS), Université de Sherbrooke , 3001 12e Avenue Nord, Sherbrooke, Québec J1H5N4, Canada
| |
Collapse
|
13
|
Auger electron-emitting 111 In-DTPA-NLS-CSL360 radioimmunoconjugates are cytotoxic to human acute myeloid leukemia (AML) cells displaying the CD123 + /CD131 − phenotype of leukemia stem cells. Appl Radiat Isot 2016; 110:1-7. [DOI: 10.1016/j.apradiso.2015.12.043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 11/18/2015] [Accepted: 12/14/2015] [Indexed: 11/19/2022]
|
14
|
Trastuzumab Labeled to High Specific Activity with 111In by Conjugation to G4 PAMAM Dendrimers Derivatized with Multiple DTPA Chelators Exhibits Increased Cytotoxic Potency on HER2-Positive Breast Cancer Cells. Pharm Res 2013; 30:1999-2009. [DOI: 10.1007/s11095-013-1044-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Accepted: 04/01/2013] [Indexed: 10/26/2022]
|
15
|
Cell-penetrating properties of the transactivator of transcription and polyarginine (R9) peptides, their conjugative effect on nanoparticles and the prospect of conjugation with arsenic trioxide. Anticancer Drugs 2012; 23:471-82. [PMID: 22241171 DOI: 10.1097/cad.0b013e32835065ed] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cell-penetrating peptides (CPPs) are short chains of amino acids with the distinct ability to cross cell plasma membranes. They are usually between seven and 30 residues in length. The mechanism of action is still a highly debated subject among researchers; it seems that a commonality between all CPPs is the presence of positively charged residues within the amino acid chain. Polyarginine and the transactivator of transcription peptide are two widely used CPPs. One distinct application of these CPPs is the ability to further enhance the therapeutic properties of a range of different agents. One group of agents of particular importance are nanoparticles (NPs). Most NPs have no mechanism for cellular uptake. Hence, by conjugating CPPs to NPs, the amount of NPs taken up by cells can be increased, and therefore, the therapeutic benefits can be maximized. Some examples of this will be explored further in this review. In addition to CPPs, the concept of conjugation with the anticancer drug arsenic trioxide is reviewed and the prospect of transactivator of transcription-conjugated arsenic trioxide albumin microspheres is also discussed. Recent locked nucleic acid technology to stabilize nucleotides (RNA or DNA) aptamer complexes able to target cancer cells more specifically and selectively to kill tumour cells and spare normal body cells. NPs tagged with modified locked nucleic acid-aptamers have the potential to kill cancer cells more specifically and effectively while sparing normal cells.
Collapse
|
16
|
111In-Bn-DTPA-nimotuzumab with/without modification with nuclear translocation sequence (NLS) peptides: an Auger electron-emitting radioimmunotherapeutic agent for EGFR-positive and trastuzumab (Herceptin)-resistant breast cancer. Breast Cancer Res Treat 2012; 135:189-200. [DOI: 10.1007/s10549-012-2137-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 06/09/2012] [Indexed: 01/25/2023]
|
17
|
Bitler BG, Schroeder JA. Anti-cancer therapies that utilize cell penetrating peptides. Recent Pat Anticancer Drug Discov 2010; 5:99-108. [PMID: 19961434 DOI: 10.2174/157489210790936252] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2008] [Accepted: 08/27/2009] [Indexed: 12/22/2022]
Abstract
Cell penetrating peptides (CPPs) are 9-35mer cationic and/or amphipathic peptides that are rapidly internalized across cell membranes. Importantly, they can be linked to a variety of cargo, including anti-cancer therapeutics, making CPPs an efficient, effective and non-toxic mechanism for drug delivery. In this review, we discuss a number of CPP conjugated therapies (CTTs) that are either patented are in the progress of patenting, and show strong promise for clinical efficacy. The CTTs discussed here target a number of different processes specific to cancer progression, including proliferation, survival and migration. In addition, many of these CTTs also increase sensitivity to current anti-cancer therapy modalities, including radiation and other DNA damaging chemotherapies, thereby decreasing the toxic dosage required for effective treatment. Mechanistically, these CTTs function in a dominant-negative manner by blocking tumor-specific protein-protein interactions with the CPP-conjugated peptide or protein. The treatment of both cell lines and mouse models demonstrates that this method of molecular targeting results in equal if not greater efficacy than current standards of care, including DNA damaging agents and topoisomerase inhibitors. For the treatment of invasive carcinoma, these CTTs have significant clinical potential to deliver highly targeted therapies without sacrificing the patient's quality of life.
Collapse
|
18
|
Kersemans V, Cornelissen B. Targeting the Tumour: Cell Penetrating Peptides for Molecular Imaging and Radiotherapy. Pharmaceuticals (Basel) 2010; 3:600-620. [PMID: 27713270 PMCID: PMC4033971 DOI: 10.3390/ph3030600] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2009] [Revised: 02/02/2010] [Accepted: 03/10/2010] [Indexed: 11/16/2022] Open
Abstract
Over the last couple of years, the number of original papers and reviews discussing various applications of cell penetrating peptides (CPPs) has grown exponentially. This is not remarkable since CPPs are capable of transporting the most varying cargo across cell membranes which is one of the biggest problems in drug delivery and targeted therapy. In this review, we focus on the use of CPPs and related peptides for delivery of imaging contrast agents and radionuclides to cells and tissues with the ultimate goal of in vivo molecular imaging and molecular radiotherapy of intracellular and even intranuclear targets.
Collapse
Affiliation(s)
- Veerle Kersemans
- Gray Institute for Radiation Oncology and Biology, University of Oxford/Old Road Campus Research Building, Off Roosevelt Drive, Churchill Hospital, Oxford OX3 7DQ, UK.
| | - Bart Cornelissen
- Gray Institute for Radiation Oncology and Biology, University of Oxford/Old Road Campus Research Building, Off Roosevelt Drive, Churchill Hospital, Oxford OX3 7DQ, UK.
| |
Collapse
|
19
|
Lee H, Hoang B, Fonge H, Reilly RM, Allen C. In vivo distribution of polymeric nanoparticles at the whole-body, tumor, and cellular levels. Pharm Res 2010; 27:2343-55. [PMID: 20195708 DOI: 10.1007/s11095-010-0068-z] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Accepted: 01/11/2010] [Indexed: 10/19/2022]
Abstract
PURPOSE Block copolymer micelles (BCMs) were functionalized with indium-111 and/or epidermal growth factor (EGF), which enabled investigation of the in vivo transport of passively and actively targeted BCMs. The integration of conventional and image-based techniques afforded novel quantitative means to achieve an in-depth insight into the fate of polymeric nanoparticles in vivo. METHODS Pharmacokinetics and biodistribution studies were performed in athymic mice bearing human breast xenografts to evaluate the whole-body transport of NT-BCMs (non-targeted, EGF-) and T-BCMs (targeted, EGF+). The intratumoral distribution of BCMs was investigated using MicroSPECT/CT and autoradiographic imaging, complemented with quantitative MATLAB® analyses. Tumors were fractionated for quantifying intracellular uptake of BCMs via γ-counting. RESULTS The intratumoral distribution of NT-BCMs and T-BCMs were found to be heterogeneous, and positively correlated with tumor vascularization (r>0.68 ± 0.04). The enhanced in vivo cell uptake and cell membrane binding of T-BCMs were found to delay their clearance from tumors overexpressing EGFR, and therefore resulted in enhanced tumor accumulation for the T-BCMs in comparison to the NT-BCMs. CONCLUSIONS Adequate passive targeting is required in order to achieve effective active targeting. Tumor physiology has a significant impact on the transvascular and intratumoral transport of passively and actively targeted BCMs.
Collapse
Affiliation(s)
- Helen Lee
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St., Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
20
|
Sawant R, Torchilin V. Intracellulartransduction using cell-penetrating peptides. ACTA ACUST UNITED AC 2010; 6:628-40. [DOI: 10.1039/b916297f] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
21
|
Fonge H, Lee H, Reilly RM, Allen C. Multifunctional Block Copolymer Micelles for the Delivery of 111In to EGFR-Positive Breast Cancer Cells for Targeted Auger Electron Radiotherapy. Mol Pharm 2009; 7:177-86. [DOI: 10.1021/mp900201v] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Humphrey Fonge
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada, Department of Medical Imaging, University of Toronto, Toronto, ON, Canada, Toronto General Research Institute, University Health Network, Toronto, ON, Canada, Department of Chemistry, University of Toronto, Toronto, ON, Canada, and STTARR Innovation Centre, Radiation Medicine Program, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada
| | - Helen Lee
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada, Department of Medical Imaging, University of Toronto, Toronto, ON, Canada, Toronto General Research Institute, University Health Network, Toronto, ON, Canada, Department of Chemistry, University of Toronto, Toronto, ON, Canada, and STTARR Innovation Centre, Radiation Medicine Program, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada
| | - Raymond M. Reilly
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada, Department of Medical Imaging, University of Toronto, Toronto, ON, Canada, Toronto General Research Institute, University Health Network, Toronto, ON, Canada, Department of Chemistry, University of Toronto, Toronto, ON, Canada, and STTARR Innovation Centre, Radiation Medicine Program, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada, Department of Medical Imaging, University of Toronto, Toronto, ON, Canada, Toronto General Research Institute, University Health Network, Toronto, ON, Canada, Department of Chemistry, University of Toronto, Toronto, ON, Canada, and STTARR Innovation Centre, Radiation Medicine Program, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada
| |
Collapse
|
22
|
Cornelissen B, Kersemans V, McLarty K, Tran L, Vallis KA, Reilly RM. In vivo monitoring of intranuclear p27(kip1) protein expression in breast cancer cells during trastuzumab (Herceptin) therapy. Nucl Med Biol 2009; 36:811-9. [PMID: 19720293 DOI: 10.1016/j.nucmedbio.2009.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 04/28/2009] [Accepted: 05/10/2009] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Trastuzumab, a humanized antibody directed against the Her2 receptor, induces the expression of p27(kip1), an intranuclear cyclin-dependent kinase inhibitor in some breast cancer cells. The aim of this study was to develop a radioimmunoconjugate (RIC) to monitor trastuzumab-induced p27(kip1) protein up-regulation in vivo. MATERIALS AND METHODS Anti-p27(kip1) IgG was purified, and conjugated to diethylenetriaminopentaacetate, to allow radiolabeling with (111)In for in vivo detection. Then tat peptide (GRKKRRQRRRPPQGYG), containing a nuclear localization sequence (underlined), was conjugated to the Fc-domain of IgG, using NaIO(4) oxidation of carbohydrates and the resulting Schiff base stabilized with NaCNBH(3). The conjugate was radiolabeled with (111)In, yielding [(111)In]-anti-p27(kip1)-tat. (111)In labeling efficiency, purity and p27(kip1) binding were measured. Trastuzumab-induced p27(kip1) up-regulation was assessed in a panel of breast cancer cell lines by Western blot analysis. Uptake and retention of [(111)In]-anti-p27(kip1)-tat were measured in MDA-MB-361 and SKBr3 cells after exposure to trastuzumab. Uptake of [(111)In]-anti-p27(kip1)-tat was determined at 72 h postintravenous injection in MDA-MB-361 xenografts in athymic mice treated with trastuzumab or saline. RESULTS [(111)In]-anti-p27(kip1)-tat was synthesized to 97% purity. The RIC was able to bind to p27(kip1) protein and internalized in the cells and was transported to the nuclei of MDA-MB-361 cells. The level of p27(kip1) protein in MDA-MB-361 cells was increased after exposure to clinically relevant doses of trastuzumab for 3 days. Trastuzumab-mediated induction of p27(kip1) was not associated with increased cellular uptake or nuclear localization of [(111)In]-anti-p27(kip1)-tat (6.53+/-0.61% vs. 6.98+/-1.36% internalized into trastuzumab-treated vs. control cells, respectively). However, retention of [(111)In]-anti-p27(kip1)-tat at 72 h was increased approximately twofold (13.5+/-1.3% vs. 6.6+/-0.6% of internalized [(111)In]-anti-p27(kip1)-tat was retained in trastuzumab-treated vs. control cells, respectively; P=.016). Immunohistochemistry showed up-regulation of p27(kip1) in trastuzumab-treated xenografts. Tumour uptake of [(111)In]-anti-p27(kip1)-tat was significantly higher in trastuzumab-treated compared to control animals (6.5+/-0.9 vs. 4.8+/-0.1 %ID/g at 72 h postinjection, respectively; P=.0065). CONCLUSION [(111)In]-Anti-p27(kip1)-tat may be useful for monitoring changes in the expression of the intranuclear protein, p27(kip1). Up-regulation of p27(kip1) resulted in increased retention of [(111)In]-anti-p27(kip1)-tat in cells treated with trastuzumab. Modest, but statistically significantly higher, retention was also observed in tumours in mice treated with trastuzumab. Since responsiveness to trastuzumab correlated to up-regulation of p27(kip1), it may be possible to use [(111)In]-anti-p27(kip1)-tat to guide treatment with Herceptin and other drugs which alter p27(kip1) expression.
Collapse
Affiliation(s)
- Bart Cornelissen
- Division of Nuclear Medicine, University Health Network, Toronto, ON, Canada M5S 3E2.
| | | | | | | | | | | |
Collapse
|
23
|
Cornelissen B, Kersemans V, McLarty K, Tran L, Reilly RM. 111In-Labeled Immunoconjugates (ICs) Bispecific for the Epidermal Growth Factor Receptor (EGFR) and Cyclin-Dependent Kinase Inhibitor, p27Kip1. Cancer Biother Radiopharm 2009; 24:163-73. [DOI: 10.1089/cbr.2008.0553] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Affiliation(s)
- Bart Cornelissen
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Veerle Kersemans
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Kristin McLarty
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Lara Tran
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Raymond M. Reilly
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
- Toronto General Research Institute, University Health Network Toronto, Ontario, Canada
| |
Collapse
|
24
|
Costantini DL, Bateman K, McLarty K, Vallis KA, Reilly RM. Trastuzumab-resistant breast cancer cells remain sensitive to the auger electron-emitting radiotherapeutic agent 111In-NLS-trastuzumab and are radiosensitized by methotrexate. J Nucl Med 2008; 49:1498-505. [PMID: 18703606 DOI: 10.2967/jnumed.108.051771] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
UNLABELLED Our goals in this study were to determine whether (111)In-trastuzumab coupled to peptides harboring nuclear localizing sequences (NLSs) could kill trastuzumab-resistant breast cancer cell lines through the emission of Auger electrons and whether the combination of radiosensitization with methotrexate (MTX) would augment the cytotoxicity of this radiopharmaceutical. METHODS Trastuzumab was derivatized with sulfosuccinimidyl-4-(N-maleimidomethyl)cyclohexane-1-carboxylate for reaction with NLS peptides and then conjugated with diethylenetriaminepentaacetic acid for labeling with (111)In. HER2 expression was determined by Western blot and by radioligand binding assay using (111)In-trastuzumab in a panel of breast cancer cell lines, including SK-BR-3, MDA-MB-231 and its HER2-transfected subclone (231-H2N), and 2 trastuzumab-resistant variants (TrR1 and TrR2). Nuclear importation of (111)In-NLS-trastuzumab and (111)In-trastuzumab in breast cancer cells was measured by subcellular fractionation, and the clonogenic survival of these cells was determined after incubation with (111)In-NLS-trastuzumab, (111)In-trastuzumab, or trastuzumab (combined with or without MTX). Survival curves were analyzed according to the dose-response model, and the radiation-enhancement ratio was calculated from the survival curve parameters. RESULTS The expression of HER2 was highest in SK-BR-3 cells (12.6 x 10(5) receptors/cell), compared with 231-H2N and TrR1 cells (6.1 x 10(5) and 5.1 x 10(5) receptors/cell, respectively), and lowest in MDA-MB-231 and TrR2 cells (0.4 x 10(5) and 0.6 x 10(5) receptors/cell, respectively). NLS peptides increased the nuclear uptake of (111)In-trastuzumab in MDA-MB-231, 231-H2N, TrR1, and TrR2 cells from 0.1%+/-0.01%, 2.5%+/-0.2%, 2.8%+/-0.7%, and 0.5%+/-0.1% to 0.5%+/-0.1%, 4.6%+/-0.1%, 5.2%+/-0.6%, and 1.5%+/-0.2%, respectively. The cytotoxicity of (111)In-NLS-trastuzumab on breast cancer cells was directly correlated with the HER2 expression densities of the cells. On a molar concentration basis, the effective concentration required to kill 50% of 231-H2N and TrR1 cells for (111)In-NLS-trastuzumab was 9- to 12-fold lower than for (111)In-trastuzumab and 16- to 77-fold lower than for trastuzumab. MDA-MB-231 and TrR2 cells were less sensitive to (111)In-NLS-trastuzumab or (111)In-trastuzumab, and both cell lines were completely insensitive to trastuzumab. The radiation-enhancement ratio induced by MTX for 231-H2N and TrR1 cells after exposure to (111)In-NLS-trastuzumab was 1.42 and 1.68, respectively. CONCLUSION Targeted Auger electron radioimmunotherapy with (111)In-NLS-trastuzumab can overcome resistance to trastuzumab, and MTX can potently enhance the sensitivity of HER2-overexpressing breast cancer cells to the lethal Auger electrons emitted by this radiopharmaceutical.
Collapse
Affiliation(s)
- Danny L Costantini
- Leslie Dan Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
25
|
Therapy of human carcinoma xenografts with antibodies to EGFr and HER-2 conjugated to radionuclides emitting low-energy electrons. Eur J Nucl Med Mol Imaging 2008; 35:1249-58. [PMID: 18265976 DOI: 10.1007/s00259-008-0731-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2007] [Accepted: 01/15/2008] [Indexed: 12/31/2022]
Abstract
PURPOSE Low-energy electrons (10-50 keV) can be effective and specific cytotoxic agents when delivered to the cell surface by antibodies, because their path length in tissue is comparable to a cell diameter. In this study, we have begun to evaluate the therapeutic potential of antibodies (Abs) conjugated to (111)In against carcinoma xenografts in nude mice. METHODS Abs to EGFr or HER-2 were labeled with (111)In to a high specific activity of approximately 1.48 GBq/mg (40 mCi/mg). They were injected into nude mice 5-6 days after inoculation of human carcinoma cells, either A431 or SK-OV-3, and tumor growth was monitored. In preliminary in vitro experiments, we calculated the cumulative decays per cell, estimated the centigray dose delivered to the nucleus, and related this to the fraction surviving. RESULTS Abs to both antigens provided significant protection in nude mouse xenograft models (p values ranging from <0.05 to <0.001). Some mice appeared to be cured, but most had delayed tumor growth. The specificity of the effect was demonstrated by testing non-reactive Abs labeled in the same way. The radioactivity was required, because unconjugated Abs had no therapeutic effect. The maximum tolerated dose was required in order for therapy to be effective, but most of the treated mice had no significant weight loss or other overt signs of toxicity. CONCLUSION Abs labeled with nuclides emitting low-energy electrons, such as (111)In, can be effective therapeutic agents against microscopic s.c. tumors. This strategy should be considered for clinical applications.
Collapse
|
26
|
Costantini DL, Hu M, Reilly RM. Update:Peptide Motifs for Insertion of Radiolabeled Biomolecules into Cells and Routing to the Nucleus for Cancer Imaging or Radiotherapeutic Applications. Cancer Biother Radiopharm 2008; 23:3-24. [DOI: 10.1089/cbr.2007.0430] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Danny L. Costantini
- Leslie Dan Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Meiduo Hu
- Leslie Dan Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Raymond M. Reilly
- Leslie Dan Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
27
|
Cornelissen B, McLarty K, Kersemans V, Scollard DA, Reilly RM. Properties of [111In]-labeled HIV-1 tat peptide radioimmunoconjugates in tumor-bearing mice following intravenous or intratumoral injection. Nucl Med Biol 2008; 35:101-10. [DOI: 10.1016/j.nucmedbio.2007.09.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Revised: 08/25/2007] [Accepted: 09/28/2007] [Indexed: 11/27/2022]
|
28
|
Hu M, Scollard D, Chan C, Chen P, Vallis K, Reilly RM. Effect of the EGFR density of breast cancer cells on nuclear importation, in vitro cytotoxicity, and tumor and normal-tissue uptake of [111In]DTPA-hEGF. Nucl Med Biol 2007; 34:887-96. [PMID: 17998090 DOI: 10.1016/j.nucmedbio.2007.06.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Revised: 06/15/2007] [Accepted: 06/25/2007] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Our objective was to evaluate the effect of epidermal growth factor receptor(s) (EGFR) density on the importation and nuclear localization of 111In-labeled diethylenetriaminepentaacetic acid human epidermal growth factor ([111In]DTPA-hEGF) in breast cancer (BC) cells in vitro and in tumor xenografts and normal tissues in vivo in athymic mice, as well as on its cytotoxicity and tumor and normal-tissue distribution. METHODS The internalization and nuclear importation of [111In]DTPA-hEGF were measured in MCF-7, MDA-MB-231, BT-474 and MDA-MB-468 BC cells (10(4), 2 x 10(5), 6 x 10(5) and 10(6) EGFR/cell, respectively). The molecular size (Mr) distribution and immunoreactivity of nuclear radioactivity were characterized. Tumor and normal-tissue uptake of [111In]DTPA-hEGF in athymic mice implanted subcutaneously with BC xenografts were compared. Nuclear radioactivity in the tumor, lungs, liver, kidneys, spleen and colon was measured. RESULTS There was a direct association between EGFR density and the nuclear localization of [111In]DTPA-hEGF in BC cells; nuclear importation approached saturation at 6 x 10(5) EGFR/cell. Almost all nuclear radioactivities exhibited an Mr of >100 kDa; immunoreactivity with anti-hEGF, anti-EGFR and anti-importin beta 1 antibodies was detected. The efflux of nuclear radioactivity was slowest for MDA-MB-468 cells. Cytotoxicity was correlated with EGFR expression. Uptake was greater in MDA-MB-468 than in MCF-7 xenografts and improved with preinjection of a 100-fold excess of unlabeled DTPA-hEGF. Nuclear importation was higher in liver, kidney and spleen cells than in tumor cells. CONCLUSION [111In]DTPA-hEGF is translocated to the nucleus of BC cells complexed with EGFR and importin beta1. Nuclear importation and cytotoxicity are effected by EGFR density. The absence of hepatic and renal toxicities in [111In]DTPA-hEGF cannot be explained by a low efficiency of nuclear importation.
Collapse
Affiliation(s)
- Meiduo Hu
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada M5S 3M2
| | | | | | | | | | | |
Collapse
|
29
|
Cornelissen B, Hu M, McLarty K, Costantini D, Reilly RM. Cellular penetration and nuclear importation properties of 111In-labeled and 123I-labeled HIV-1 tat peptide immunoconjugates in BT-474 human breast cancer cells. Nucl Med Biol 2007; 34:37-46. [PMID: 17210460 DOI: 10.1016/j.nucmedbio.2006.10.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2006] [Revised: 10/26/2006] [Accepted: 10/30/2006] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Our objective was to compare the cell penetration and nuclear importation properties of 111In-labeled and 123I-labeled immunoconjugates (ICs) composed of 16-mer peptides (GRKKRRQRRRPPQGYG) derived from HIV-1 transactivator of transcription (tat) protein and anti-mouse IgG (mIgG) in BT-474 breast cancer (BC) cells. METHODS [111In]tat ICs were constructed by site-specific conjugation of tat peptides to NaIO4(-)-oxidized carbohydrates in the Fc domain of diethylenetriaminepentaacetic-acid-modified anti-mIgG antibodies. Immunoreactivity against mIgG was assessed in a competition assay. The kinetics of the accumulation of [111In]anti-mIgG-tat IC and [123I]anti-mIgG-tat ICs in BT-474 cells and the elimination of radioactivity from cells, cytoplasm or nuclei were determined. The effects of excess tat peptides or NH4Cl (an inhibitor of endosomal acidification) on cellular uptake and nuclear importation of [111In]anti-mIgG-tat were measured. RESULTS [111In]anti-mIgG-tat was >97% radiochemically pure and exhibited preserved immunoreactivity with mIgG epitopes. [123I]Anti-mIgG-tat penetrated BT-474 cells more rapidly than [111In]anti-mIgG-tat ICs and achieved a 1.5-fold to a 2-fold higher uptake in cells and nuclei. Cell penetration and nuclear uptake of [111In]anti-mIgG-tat were inhibited by excess tat peptides and NH4Cl. Elimination of radioactivity from BT-474 cells and nuclei was more rapid and complete for 123I-labeled than for 111In-labeled anti-mIgG-tat ICs. CONCLUSION Tat peptides derived from HIV-1 tat protein promoted the penetration and nuclear uptake of radioactivity following the incubation of 111In-labeled and 123I-labeled anti-mIgG antibodies with BT-474 human BC cells. 111In-labeled tat ICs are feasible for inserting radionuclides into cancer cells with potential for targeting intracellular and, particularly, nuclear epitopes for imaging and/or radiotherapeutic applications.
Collapse
Affiliation(s)
- Bart Cornelissen
- Division of Nuclear Medicine, University Health Network, Toronto, ON, Canada M5S 3M2
| | | | | | | | | |
Collapse
|
30
|
McLarty K, Reilly RM. Molecular Imaging as a Tool for Personalized and Targeted Anticancer Therapy. Clin Pharmacol Ther 2007; 81:420-4. [PMID: 17339871 DOI: 10.1038/sj.clpt.6100096] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- K McLarty
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
31
|
Hu M, Chen P, Wang J, Scollard DA, Vallis KA, Reilly RM. 123I-labeled HIV-1 tat peptide radioimmunoconjugates are imported into the nucleus of human breast cancer cells and functionally interact in vitro and in vivo with the cyclin-dependent kinase inhibitor, p21WAF-1/Cip-1. Eur J Nucl Med Mol Imaging 2006; 34:368-77. [PMID: 17021818 DOI: 10.1007/s00259-006-0189-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2005] [Accepted: 06/09/2006] [Indexed: 11/26/2022]
Abstract
PURPOSE To evaluate the internalization and nuclear translocation of (123)I-tat-peptide radioimmunoconjugates in MDA-MB-468 breast cancer cells and their ability to interact with the cyclin-dependent kinase inhibitor, p21(WAF-1/Cip-1). METHODS Peptides [GRKKRRQRRRPPQGYGC] harboring the nuclear-localizing sequence from HIV tat domain were conjugated to anti-p21(WAF-1/Cip-1) antibodies. Immunoreactivity was assessed by Western blot using lysate from MDA-MB-468 cells exposed to EGF to induce p21(WAF-1/Cip-1). Internalization and nuclear translocation were measured. The ability of tat-anti-p21(WAF-1/Cip-1) to block G(1)-S phase arrest in MDA-MB-468 cells caused by EGF-induced p21(WAF-1/Cip-1) was evaluated. Tumor and normal tissue uptake were determined at 48 h p.i. in athymic mice implanted s.c. with MDA-MB-468 xenografts injected intratumorally with EGF. RESULTS There was 13.4+/-0.2% of radioactivity internalized by MDA-MB-468 cells incubated with (123)I-tat-anti-p21(WAF-1/Cip-1) and 34.6+/-3.1% imported into the nucleus. Tat-anti-p21(WAF-1/Cip-1)(8 muM) decreased the proportion of EGF-treated cells in G(1) phase from 81.9+/-0.7% to 46.1+/-0.7% (p<0.001), almost restoring the G(1) phase fraction to that of unexposed cells (25.8+/-0.2%). Non-specific tat-mouse IgG did not block EGF-induced G(1)-S phase arrest. Tumor uptake of radioactivity was higher in mice injected with EGF to induce p21(WAF-1/Cip-1) than in mice not receiving EGF (3.1+/-0.4% versus 1.8+/-0.2% ID/g; p=0.04). Western blot analysis of tumors revealed a threefold increase in the p21(WAF-1/Cip-1)/beta-actin ratio. CONCLUSION We conclude that intracellular and nuclear epitopes in cancer cells can be functionally targeted with tat-radioimmunoconjugates to exploit many more epitopes for imaging and radiotherapeutic applications than have previously been accessible.
Collapse
Affiliation(s)
- Meiduo Hu
- Division of Nuclear Medicine, University Health Network, Toronto, ON, Canada
| | | | | | | | | | | |
Collapse
|