1
|
Buck J, Perez‐Balderas F, Zarghami N, Johanssen V, Khrapitchev AA, Larkin JR, Sibson NR. Imaging angiogenesis in an intracerebrally induced model of brain macrometastasis using α v β 3 -targeted iron oxide microparticles. NMR IN BIOMEDICINE 2023; 36:e4948. [PMID: 37038086 PMCID: PMC10909432 DOI: 10.1002/nbm.4948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 06/19/2023]
Abstract
Brain metastasis is responsible for a large proportion of cancer mortality, and there are currently no effective treatments. Moreover, the impact of treatments, particularly antiangiogenic therapeutics, is difficult to ascertain using current magnetic resonance imaging (MRI) methods. Imaging of the angiogenic vasculature has been successfully carried out in solid tumours using microparticles of iron oxide (MPIO) conjugated to a Arg-Gly-Asp peptide (RGD) targeting integrin αv β3 . The aim of this study was to determine whether RGD-MPIO could be used to identify angiogenic blood vessels in brain metastases in vivo. A mouse model of intracerebrally implanted brain macrometastasis was established through intracerebral injection of 4T1-GFP cells. T2 *-weighted imaging was used to visualise MPIO-induced hypointense voxels in vivo, and Prussian blue staining was used to visualise MPIO and endogenous iron histologically ex vivo. The RGD-MPIO showed target-specific binding in vivo, but the sensitivity of the agent for visualising angiogenic vessels per se was reduced by the presence of endogenous iron-laden macrophages in larger metastases, resulting in pre-existing hypointense areas within the tumour. Further, our data suggest that peptide-targeted MPIO, but not antibody-targeted MPIO, are taken up by perivascular macrophages within the macrometastatic microenvironment, resulting in additional nonspecific contrast. While pre-MPIO imaging will circumvent the issues surrounding pre-existing hypointensities and enable detection of specific contrast, our preliminary findings suggest that the use of antibodies rather than peptides as the targeting ligand may represent a preferable route forward for new angiogenesis-targeted molecular MRI agents.
Collapse
Affiliation(s)
- Jessica Buck
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Francisco Perez‐Balderas
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Niloufar Zarghami
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Vanessa Johanssen
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Alexandre A. Khrapitchev
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - James R. Larkin
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Nicola R. Sibson
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| |
Collapse
|
2
|
Braun D, Judmann B, Cheng X, Wängler B, Schirrmacher R, Fricker G, Wängler C. Synthesis, Radiolabeling, and In Vitro and In Vivo Characterization of Heterobivalent Peptidic Agents for Bispecific EGFR and Integrin α vβ 3 Targeting. ACS OMEGA 2023; 8:2793-2807. [PMID: 36687076 PMCID: PMC9850772 DOI: 10.1021/acsomega.2c07484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 12/15/2022] [Indexed: 06/17/2023]
Abstract
Radiolabeled heterobivalent peptidic ligands (HBPLs) are a highly promising compound class for the sensitive and specific visualization of tumors as they often exhibit superior properties compared to their monospecific counterparts and are able to concomitantly or complementarily address different receptor types. The combination of two receptor-specific agents targeting the epidermal growth factor receptor (EGFR) and the integrin αvβ3 in one HBPL would constitute a synergistic combination of binding motifs as these two receptor types are concurrently overexpressed on several human tumor types and are closely associated with disease progression and metastasis. Here, we designed and synthesized two heterobivalent radioligands consisting of the EGFR-specific peptide GE11 and αvβ3-specific cyclic RGD peptides, bearing a (1,4,7-triazacyclononane-4,7-diyl)diacetic acid-1-glutaric acid chelator for efficient radiolabeling and linkers of different lengths between both peptides. Both HBPLs were radiolabeled with 68Ga3+ in high radiochemical yields, purities of 96-99%, and molar activities of 36-88 GBq/μmol. [68Ga]Ga-1 and [68Ga]Ga-2 were evaluated for their log D(7.4) and stability toward degradation by human serum peptidases, showing a high hydrophilicity for both agents of -3.07 ± 0.01 and -3.44 ± 0.08 as well as a high stability toward peptidase degradation in human serum with half-lives of 272 and 237 min, respectively. Further on, the in vitro receptor binding profiles of both HBPLs to the target EGF and integrin αvβ3 receptors were assessed on EGFR-positive A431 and αvβ3-positive U87MG cells. Finally, we investigated the in vivo pharmacokinetics of HBPL [68Ga]Ga-1 by positron emission tomography/computed tomography imaging in A431 tumor-bearing xenograft mice to assess its potential for the receptor-specific visualization of EGFR- and/or αvβ3-expressing tumors. In these experiments, [68Ga]Ga-1 demonstrated a tumor uptake of 2.79 ± 1.66% ID/g, being higher than in all other organs and tissues apart from kidneys and blood at 2 h p.i. Receptor blocking studies revealed the observed tumor uptake to be solely mediated by integrin αvβ3, whereas no contribution of the GE11 peptide sequence to tumor uptake via the EGFR could be determined. Thus, the approach to develop radiolabeled EGFR- and integrin αvβ3-bispecific HBPLs is in general feasible although another peptide lead structure than GE11 should be used as the basis for the EGFR-specific part of the agents.
Collapse
Affiliation(s)
- Diana Braun
- Biomedical
Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty
Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Molecular
Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine,
Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Benedikt Judmann
- Biomedical
Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty
Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Molecular
Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine,
Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Xia Cheng
- Molecular
Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine,
Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Björn Wängler
- Molecular
Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine,
Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Ralf Schirrmacher
- Department
of Oncology, Division of Oncological Imaging, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada
| | - Gert Fricker
- Institute
of Pharmacy and Molecular Biotechnology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Carmen Wängler
- Biomedical
Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty
Mannheim, Heidelberg University, 68167 Mannheim, Germany
| |
Collapse
|
3
|
Perrins RD, McCarthy LA, Robinson A, Spry KL, Cognet V, Ferreira A, Porter J, Garcίa CE, Rodriguez MÁ, Lopez D, Perera I, Conlon K, Barrientos A, Coulter T, Pace A, Hale SJM, Ferrari E, Bachrati CZ. Targeting Ultrasmall Gold Nanoparticles with cRGD Peptide Increases the Uptake and Efficacy of Cytotoxic Payload. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12224013. [PMID: 36432299 PMCID: PMC9696180 DOI: 10.3390/nano12224013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/07/2022] [Accepted: 11/14/2022] [Indexed: 05/06/2023]
Abstract
Cyclic arginyl-glycyl-aspartic acid peptide (cRGD) peptides show a high affinity towards αVβ3 integrin, a receptor overexpressed in many cancers. We aimed to combine the versatility of ultrasmall gold nanoparticles (usGNP) with the target selectivity of cRGD peptide for the directed delivery of a cytotoxic payload in a novel design. usGNPs were synthesized with a modified Brust-Schiffrin method and functionalized via amide coupling and ligand exchange and their uptake, intracellular trafficking, and toxicity were characterized. Our cRGD functionalized usGNPs demonstrated increased cellular uptake by αVβ3 integrin expressing cells, are internalized via clathrin-dependent endocytosis, accumulated in the lysosomes, and when loaded with mertansine led to increased cytotoxicity. Targeting via cRGD functionalization provides a mechanism to improve the efficacy, tolerability, and retention of therapeutic GNPs.
Collapse
Affiliation(s)
| | - Lee-Anne McCarthy
- School of Life Sciences, University of Lincoln, Joseph Banks Laboratories, Green Lane, Lincoln LN6 7DL, UK
| | - Angela Robinson
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Kelly L. Spry
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Valentin Cognet
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Avelino Ferreira
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - John Porter
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | | | | | - Diana Lopez
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Ibon Perera
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Kelly Conlon
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Africa Barrientos
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Tom Coulter
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Alessandro Pace
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Sarah J. M. Hale
- Midatech Pharma Plc, 1 Caspian Point, Caspian Way, Cardiff CF10 4DQ, UK
| | - Enrico Ferrari
- School of Life Sciences, University of Lincoln, Joseph Banks Laboratories, Green Lane, Lincoln LN6 7DL, UK
| | - Csanad Z. Bachrati
- School of Life Sciences, University of Lincoln, Joseph Banks Laboratories, Green Lane, Lincoln LN6 7DL, UK
- Correspondence: ; Tel.: +44-1522-886787
| |
Collapse
|
4
|
Damerow H, Cheng X, von Kiedrowski V, Schirrmacher R, Wängler B, Fricker G, Wängler C. Toward Optimized 89Zr-Immuno-PET: Side-by-Side Comparison of [ 89Zr]Zr-DFO-, [ 89Zr]Zr-3,4,3-(LI-1,2-HOPO)- and [ 89Zr]Zr-DFO*-Cetuximab for Tumor Imaging: Which Chelator Is the Most Suitable? Pharmaceutics 2022; 14:pharmaceutics14102114. [PMID: 36297549 PMCID: PMC9611803 DOI: 10.3390/pharmaceutics14102114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/21/2022] Open
Abstract
89Zr represents a highly favorable positron emitter for application in immuno-PET (Positron Emission Tomography) imaging. Clinically, the 89Zr4+ ion is introduced into antibodies by complexation with desferrioxamine B. However, producing complexes of limited kinetic inertness. Therefore, several new chelators for 89Zr introduction have been developed over the last years. Of these, the direct comparison of the most relevant ones for clinical translation, DFO* and 3,4,3-(LI-1,2-HOPO), is still missing. Thus, we directly compared DFO with DFO* and 3,4,3-(LI-1,2-HOPO) immunoconjugates to identify the most suitable agent stable 89Zr-complexation. The chelators were introduced into cetuximab, and an optical analysis method was developed, enabling the efficient quantification of derivatization sites per protein. The cetuximab conjugates were efficiently obtained and radiolabeled with 89Zr at 37 °C within 30 min, giving the [89Zr]Zr-cetuximab derivatives in high radiochemical yields and purities of >99% as well as specific activities of 50 MBq/mg. The immunoreactive fraction of all 89Zr-labeled cetuximab derivatives was determined to be in the range of 86.5−88.1%. In vivo PET imaging and ex vivo biodistribution studies in tumor-bearing animals revealed a comparable and significantly higher kinetic inertness for both [89Zr]Zr-3,4,3-(LI-1,2-HOPO)-cetuximab and [89Zr]Zr-DFO*-cetuximab, compared to [89Zr]Zr-DFO-cetuximab. Of these, [89Zr]Zr-DFO*-cetuximab showed a considerably more favorable pharmacokinetic profile with significantly lower liver and spleen retention than [89Zr]Zr-3,4,3-(LI-1,2-HOPO)-cetuximab. Since [89Zr]Zr-DFO* demonstrates a very high kinetic inertness, paired with a highly favorable pharmacokinetic profile of the resulting antibody conjugate, DFO* currently represents the most suitable chelator candidate for stable 89Zr-radiolabeling of antibodies and clinical translation.
Collapse
Affiliation(s)
- Helen Damerow
- Biomedical Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Xia Cheng
- Molecular Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Valeska von Kiedrowski
- Molecular Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Ralf Schirrmacher
- Division of Oncologic Imaging, Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Björn Wängler
- Molecular Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Carmen Wängler
- Biomedical Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Correspondence:
| |
Collapse
|
5
|
Judmann B, Braun D, Schirrmacher R, Wängler B, Fricker G, Wängler C. Toward the Development of GE11-Based Radioligands for Imaging of Epidermal Growth Factor Receptor-Positive Tumors. ACS OMEGA 2022; 7:27690-27702. [PMID: 35967067 PMCID: PMC9366781 DOI: 10.1021/acsomega.2c03407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/15/2022] [Indexed: 06/15/2023]
Abstract
The epidermal growth factor receptor (EGFR) is closely associated with tumor development and progression and thus an important target structure for imaging and therapy of various tumors. As a result of its important role in malignancies of various origins and the fact that antibody-based compounds targeting the EGFR have significant drawbacks in terms of in vivo pharmacokinetics, several attempts have been made within the last five years to develop peptide-based EGFR-specific radioligands based on the GE11 scaffold. However, none of these approaches have shown convincing results so far, which has been proposed to be attributed to different potential challenges associated with the GE11 lead structure: first, an aggregation of radiolabeled peptides, which might prevent their interaction with their target receptor, or second, a relatively low affinity of monomeric GE11, necessitating its conversion into a multimeric or polymeric form to achieve adequate EGFR-targeting properties. In the present work, we investigated if these aforementioned points are indeed critical and if the EGFR-targeting ability of GE11 can be improved by choosing an appropriate hydrophilic molecular design or a peptide multimer system to obtain a promising radiopeptide for the visualization of EGFR-overexpressing malignancies by positron emission tomography (PET). For this purpose, we developed several monovalent 68Ga-labeled GE11-based agents, a peptide homodimer and a homotetramer to overcome the challenges associated with GE11. The developed ligands were successfully labeled with 68Ga3+ in high radiochemical yields of ≥97% and molar activities of 41-104 GBq/μmol. The resulting radiotracers presented log D(7.4) values between -2.17 ± 0.21 and -3.79 ± 0.04 as well as a good stability in human serum with serum half-lives of 112 to 217 min for the monovalent radiopeptides and 84 and 62 min for the GE11 homodimer and homotetramer, respectively. In the following in vitro studies, none of the 68Ga-labeled radiopeptides demonstrated a considerable EGF receptor-specific uptake in EGFR-positive A431 cells. Moreover, none of the agents was able to displace [125I]I-EGF from the EGFR in competitive displacement assays in the same cell line in concentrations of up to 1 mM, whereas the endogenous receptor ligand hEGF demonstrated a high affinity of 15.2 ± 3.3 nM. These results indicate that it is not the aggregation of the GE11 sequence that seems to be the factor limiting the usefulness of the peptide as basis for radiotracer design but the limited affinity of monovalent and small homomultivalent GE11-based radiotracers to the EGFR. This highlights that the development of small-molecule GE11-based radioligands is not promising.
Collapse
Affiliation(s)
- Benedikt Judmann
- Biomedical
Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty
Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Molecular
Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine,
Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Diana Braun
- Biomedical
Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty
Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Molecular
Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine,
Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Ralf Schirrmacher
- Department
of Oncology, Division of Oncological Imaging, University of Alberta, 11560 University Avenue, T6G 1Z2 Edmonton, AB, Canada
| | - Björn Wängler
- Molecular
Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine,
Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Gert Fricker
- Institute
of Pharmacy and Molecular Biotechnology, University of Heidelberg, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany
| | - Carmen Wängler
- Biomedical
Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty
Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| |
Collapse
|
6
|
Growth and site-specific organization of micron-scale biomolecular devices on living mammalian cells. Nat Commun 2021; 12:5729. [PMID: 34593818 PMCID: PMC8484582 DOI: 10.1038/s41467-021-25890-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 08/27/2021] [Indexed: 12/11/2022] Open
Abstract
Mesoscale molecular assemblies on the cell surface, such as cilia and filopodia, integrate information, control transport and amplify signals. Designer cell-surface assemblies could control these cellular functions. Such assemblies could be constructed from synthetic components ex vivo, making it possible to form such structures using modern nanoscale self-assembly and fabrication techniques, and then oriented on the cell surface. Here we integrate synthetic devices, micron-scale DNA nanotubes, with mammalian cells by anchoring them by their ends to specific cell surface receptors. These filaments can measure shear stresses between 0-2 dyn/cm2, a regime important for cell signaling. Nanotubes can also grow while anchored to cells, thus acting as dynamic cell components. This approach to cell surface engineering, in which synthetic biomolecular assemblies are organized with existing cellular architecture, could make it possible to build new types of sensors, machines and scaffolds that can interface with, control and measure properties of cells. Mesoscale molecular assemblies on the cell surface integrate information and amplify signals. Here the authors integrate DNA nanotubes in a controlled manner with mammalian cells to act as sheer stress meters.
Collapse
|
7
|
Schneider H, Englert S, Macarrón Palacios A, Lerma Romero JA, Ali A, Avrutina O, Kolmar H. Synthetic Integrin-Targeting Dextran-Fc Hybrids Efficiently Inhibit Tumor Proliferation In Vitro. Front Chem 2021; 9:693097. [PMID: 34368077 PMCID: PMC8339797 DOI: 10.3389/fchem.2021.693097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/21/2021] [Indexed: 11/13/2022] Open
Abstract
Herein, we present the design, synthesis, and biological evaluation of novel integrin-targeting molecular hybrids combining RGD peptides and a potent cytotoxin presented on dextran polysaccharides. Based on an aglycosylated Fc as a centerpiece, endosomal-cleavable cytotoxic agent monomethyl auristatin E (MMAE) and dextran as multimerization site were covalently connected by two bioorthogonal enzyme-mediated reactions site-specifically. Decoration of dextran with cyclic RGD peptides, introduced by copper “click” reaction, resulted in the final constructs with the potential to kill integrin-overexpressing tumor cells. We found that these modifications had little impact on the stability of the Fc scaffold and the RGD-bearing construct showed good binding properties of αvβ3-expressing U87MG cells. Furthermore, the construct showed a remarkable antiproliferative activity. These results demonstrate the general capability of our design to provoke receptor-mediated endocytosis upon binding to the cellular surface, followed by endosomal cleavage of the linkage between Fc-dextran and MMAE and its subsequent release. Our approach opens new avenues to transcribe small molecule binders into tailor-made multimeric molecular hybrids with antitumor potential.
Collapse
Affiliation(s)
- Hendrik Schneider
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Germany
| | - Simon Englert
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Germany
| | - Arturo Macarrón Palacios
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Germany
| | | | - Ataurehman Ali
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Germany
| | - Olga Avrutina
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Germany
| |
Collapse
|
8
|
Li G, Zhang Q. Calibration-Free Single-Molecule Absolute Quantification Using Super-resolution Microscopy. Anal Chem 2021; 93:6195-6204. [PMID: 33826293 DOI: 10.1021/acs.analchem.1c00390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Single-molecule (SM) quantification has become a powerful analytical technique in the fields of physics, chemistry, and biology. SM imaging, especially with super-resolution (SR) techniques, has dramatically facilitated the study of individual molecules that may function as disease-related biomarkers. Although multiple properties can be used for quantitative imaging analysis, counting may be the simplest and most direct way. Consequently, how to utilize the greater spatial resolution to overcome undercounting or overcounting errors in certain conditions shows promising potential to unravel intracellular mechanisms of isolated biomolecules. From this perspective, we present an absolute quantification approach, termed crucial connected-component entropy (CCCE), with subresolution accuracy for the SR SM detection platform without the need for prior knowledge of calibration, and a cross-validation analytical pipeline based on SM profiling for nanoscale performance assessments. Considering its high efficiency, accuracy, and robustness for routine SM quantification compared with commonly used strategies, we believe that this protocol will indubitably find wide applications in biochemistry research, drug discovery, and clinical diagnostics, especially molecular diagnostics.
Collapse
Affiliation(s)
- Guang Li
- Institute of Biomedical Engineering, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, P. R. China.,The First Affiliated Hospital, Jinan University, Guangzhou 510632, P. R. China
| | - Qiqing Zhang
- Institute of Biomedical Engineering, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, P. R. China
| |
Collapse
|
9
|
Perini G, Giulimondi F, Palmieri V, Augello A, Digiacomo L, Quagliarini E, Pozzi D, Papi M, Caracciolo G. Inhibiting the Growth of 3D Brain Cancer Models with Bio-Coronated Liposomal Temozolomide. Pharmaceutics 2021; 13:pharmaceutics13030378. [PMID: 33809262 PMCID: PMC7999290 DOI: 10.3390/pharmaceutics13030378] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/03/2021] [Accepted: 03/05/2021] [Indexed: 01/16/2023] Open
Abstract
Nanoparticles (NPs) have emerged as an effective means to deliver anticancer drugs into the brain. Among various forms of NPs, liposomal temozolomide (TMZ) is the drug-of-choice for the treatment and management of brain tumours, but its therapeutic benefit is suboptimal. Although many possible reasons may account for the compromised therapeutic efficacy, the inefficient tumour penetration of liposomal TMZ can be a vital obstacle. Recently, the protein corona, i.e., the layer of plasma proteins that surround NPs after exposure to human plasma, has emerged as an endogenous trigger that mostly controls their anticancer efficacy. Exposition of particular biomolecules from the corona referred to as protein corona fingerprints (PCFs) may facilitate interactions with specific receptors of target cells, thus, promoting efficient internalization. In this work, we have synthesized a set of four TMZ-encapsulating nanomedicines made of four cationic liposome (CL) formulations with systematic changes in lipid composition and physical−chemical properties. We have demonstrated that precoating liposomal TMZ with a protein corona made of human plasma proteins can increase drug penetration in a 3D brain cancer model derived from U87 human glioblastoma multiforme cell line leading to marked inhibition of tumour growth. On the other side, by fine-tuning corona composition we have also provided experimental evidence of a non-unique effect of the corona on the tumour growth for all the complexes investigated, thus, clarifying that certain PCFs (i.e., APO-B and APO-E) enable favoured interactions with specific receptors of brain cancer cells. Reported results open new perspectives into the development of corona-coated liposomal drugs with enhanced tumour penetration and antitumour efficacy.
Collapse
Affiliation(s)
- Giordano Perini
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (G.P.); (V.P.)
- Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy;
| | - Francesca Giulimondi
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy; (F.G.); (L.D.); (D.P.)
| | - Valentina Palmieri
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (G.P.); (V.P.)
- Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy;
- Istituto dei Sistemi Complessi, CNR, Via dei Taurini 19, 00185 Rome, Italy
| | - Alberto Augello
- Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy;
| | - Luca Digiacomo
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy; (F.G.); (L.D.); (D.P.)
| | - Erica Quagliarini
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy;
| | - Daniela Pozzi
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy; (F.G.); (L.D.); (D.P.)
| | - Massimiliano Papi
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (G.P.); (V.P.)
- Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy;
- Correspondence: (M.P.); (G.C.)
| | - Giulio Caracciolo
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy; (F.G.); (L.D.); (D.P.)
- Correspondence: (M.P.); (G.C.)
| |
Collapse
|
10
|
Chen D, Ganesh S, Wang W, Lupieri A, Amiji M. Role of vitronectin-rich protein corona on tumor-specific siRNA delivery and transfection with lipid nanoparticles. Nanomedicine (Lond) 2021; 16:535-551. [PMID: 33683145 DOI: 10.2217/nnm-2020-0428] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To evaluate the role of vitronectin-enriched protein corona on systemic delivery of siRNA-encapsulated cationic lipid nanoparticles (LNPs) to αvβ3 integrin expressing solid tumors. Materials & methods: 1,2-Dioleoyl-3-trimethylammonium-propane LNPs were formulated, protein corona formed in nude mice serum and its impact on drug delivery were analyzed. Results: 1,2-Dioleoyl-3-trimethylammonium-propane-containing LNP led to enhanced recruitment of vitronectin and showed preferential transfection to αvβ3-expressed cells relative to controls. Upon systemic administration in mice, the LNPs accumulated in the αvβ3-expressing endothelial lining of the tumor blood vessels before reaching tumor cells. Conclusion: These results present an optimized LNP that selectively recruits endogenous proteins in situ to its corona which may lead to enhanced delivery and transfection in tissues of interest.
Collapse
Affiliation(s)
- Dongyu Chen
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA 02115, USA
| | | | - Weimin Wang
- Dicerna Pharmaceuticals, Lexington, MA 02421, USA
| | | | - Mansoor Amiji
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA 02115, USA.,Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
11
|
Borbély A, Thoreau F, Figueras E, Kadri M, Coll J, Boturyn D, Sewald N. Synthesis and Biological Characterization of Monomeric and Tetrameric RGD-Cryptophycin Conjugates. Chemistry 2020; 26:2602-2605. [PMID: 31943410 PMCID: PMC7064988 DOI: 10.1002/chem.201905437] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Indexed: 01/28/2023]
Abstract
The effective delivery of cytotoxic agents to tumor cells is a key challenge in anticancer therapy. Multivalent integrinspecific ligands are considered a promising tool to increase the binding affinity, selectivity, and internalization efficiency of small-molecule drug conjugates. Herein, we report the synthesis and biological evaluation of a multimeric conjugate containing the high-affinity integrin αv β3 binding ligand RAFT-c(RGDfK)4 , a lysosomally cleavable Val-Cit linker, and cryptophycin-55 glycinate, a potent inhibitor of tubulin polymerization. In vitro cytotoxicity assays verified that the multimeric RGD-cryptophycin conjugate displays improved potency compared to the monomeric analogue in integrin αv β3 overexpressing tumor cell lines, while significantly reduced activity was observed in the integrin-negative cell line.
Collapse
Affiliation(s)
- Adina Borbély
- Organic and Bioorganic ChemistryDepartment of ChemistryBielefeld UniversityUniversitätsstraße 2533615BielefeldGermany
| | - Fabien Thoreau
- CNRS, Department of Molecular ChemistryUniversity Grenoble Alpes, UMR 525038000GrenobleFrance
| | - Eduard Figueras
- Organic and Bioorganic ChemistryDepartment of ChemistryBielefeld UniversityUniversitätsstraße 2533615BielefeldGermany
| | - Malika Kadri
- Institute for Advanced BiosciencesUniversity Grenoble Alpes, INSERM U1209—UMR CNRS 530938700GrenobleFrance
| | - Jean‐Luc Coll
- Institute for Advanced BiosciencesUniversity Grenoble Alpes, INSERM U1209—UMR CNRS 530938700GrenobleFrance
| | - Didier Boturyn
- CNRS, Department of Molecular ChemistryUniversity Grenoble Alpes, UMR 525038000GrenobleFrance
| | - Norbert Sewald
- Organic and Bioorganic ChemistryDepartment of ChemistryBielefeld UniversityUniversitätsstraße 2533615BielefeldGermany
| |
Collapse
|
12
|
Kopatz V, Selzer E. Quantitative and qualitative analysis of integrin subtype expression in melanocytes and melanoma cells. J Recept Signal Transduct Res 2020; 40:237-245. [PMID: 32066306 DOI: 10.1080/10799893.2020.1727923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Objectives: Changes in the integrin expression pattern have been associated with the malignant transformation of melanocytes suggesting that integrins may be potential biomarkers as well as molecular targets for individualized therapy. Since there is a lack of comprehensive qualitative and quantitative expression data, we characterized the integrin expression profile in normal and malignant human cells of the melanocytic lineage.Methods: Seven melanoma cell lines as well as normal human melanocytes were investigated in western blots including recombinant integrin subunits for quantification.Results: Expression patterns were heterogeneous. In melanoma, overexpression of α4, α6, αL, β5, and β6 was found. Integrins α7, α9, and β4 were overexpressed in a subset of the melanoma cell lines. Overexpression was defined as a lack of expression in melanocytes but expression in more than half (4) of the melanoma lines. 1.9 to 6.7 × 106 integrin molecules (about 0.3% of total cellular protein) were estimated to be expressed per cell. Expression of integrin αE at the protein level was found in melanoma and melanocytes, to the best of our knowledge, for the first time. Integrins αM and β2 were not detected.Conclusion: Integrins α4, α6, αL, β5, and β6 appear to be overexpressed in melanoma cells. These subunits may serve as biomarkers and/or therapeutic targets.
Collapse
Affiliation(s)
- Verena Kopatz
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
| | - Edgar Selzer
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
13
|
Wang T, Li G, Wang D, Li F, Men D, Hu T, Xi Y, Zhang XE. Quantitative profiling of integrin αvβ3 on single cells with quantum dot labeling to reveal the phenotypic heterogeneity of glioblastoma. NANOSCALE 2019; 11:18224-18231. [PMID: 31560005 DOI: 10.1039/c9nr01105f] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
The distribution, localization and density of individual molecules (e.g. drug-specific receptors) on single cells can offer profound information about cell phenotypes. Profiling this information is a new research direction within the field of single cell biology, but it remains technically challenging. Through the combined use of quantum dot labeling, structured illumination microscopy (SIM) and computer-aided local surface reconstruction, we acquired a 3D imaging map of a drug target molecule, integrin αvβ3, on glioblastoma cells at the single cell level. The results revealed that integrin αvβ3 exhibits discrete distribution on the surface of glioblastoma cells, with its density differing significantly among cell lines. The density is illustrated as the approximate number of target molecules per μm2 on the irregular cell surface, ranging from 0 to 1.6. Functional studies revealed that the sensitivity of glioblastoma cells to inhibitor molecules depends on the density of the target molecules. After inhibitor treatment, the viability and invasion ability of different glioblastoma cells were highly correlated with the density of integrin αvβ3 on their surfaces. This study not only provides a novel protocol for the quantitative analysis of surface proteins from irregular single cells, but also offers a clue for understanding the heterogeneity of tumor cells on the basis of molecular phenotypes. Thus, this work has potential significance in guiding targeted therapies for cancers.
Collapse
Affiliation(s)
- Tingting Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Shaw SK, Schreiber CL, Roland FM, Battles PM, Brennan SP, Padanilam SJ, Smith BD. High expression of integrin αvβ3 enables uptake of targeted fluorescent probes into ovarian cancer cells and tumors. Bioorg Med Chem 2018; 26:2085-2091. [PMID: 29548784 DOI: 10.1016/j.bmc.2018.03.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/04/2018] [Accepted: 03/05/2018] [Indexed: 02/06/2023]
Abstract
The cell line OVCAR-4 was recently ranked as one of the most representative cell lines for high grade serous ovarian cancer (HGSOC). However, little work has been done to assess the susceptibility of OVCAR-4 cells and tumors to the more common types of molecular targeting. Proteome profiles suggest OVCAR-4 express high levels of integrin αvβ3 receptors. Using flow cytometry with fluorescent antibodies we determined that OVCAR-4 cells have high number of integrin αvβ3 receptors ([9.8 ± 2.5] × 104/cell) compared to the well-characterized cell line U87-MG ([5.2 ± 1.4] × 104/cell). However, OVCAR-4 cells also have roughly three times the surface area of U87-MG cells, so the average αvβ3 receptor density is actually lower (11 ± 3 versus 18 ± 6 receptors/µm2). A series of new fluorescent molecular probes was prepared with structures comprised of a deep-red squaraine fluorophore (∼680 nm emission) covalently attached to zero, one, or two cyclic pentapeptide cRGD sequences for integrin targeting. Microscopy studies showed that uptake of the divalent probe into cultured OVCAR-4 cells was 2.2 ± 0.4 higher than the monovalent probe, which in turn was 2.2 ± 0.4 higher than the untargeted probe. This probe targeting trend was also seen in OVCAR-4 mouse tumor models. The results suggest that clinically relevant OVCAR-4 cells can be targeted using molecular probes based on αvβ3 integrin receptor antagonists such as the cRGD peptide. Furthermore, deep-red fluorescent cRGD-squaraine probes have potential as targeted stains of cancerous tissue associated with HGSOC in surgery and pathology settings.
Collapse
Affiliation(s)
- Scott K Shaw
- University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN 46545, United States
| | - Cynthia L Schreiber
- University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN 46545, United States
| | - Felicia M Roland
- University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN 46545, United States
| | - Paul M Battles
- University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN 46545, United States
| | - Seamus P Brennan
- University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN 46545, United States
| | - Simon J Padanilam
- University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN 46545, United States
| | - Bradley D Smith
- University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN 46545, United States.
| |
Collapse
|
15
|
Bitsouni V, Trucu D, Chaplain MAJ, Eftimie R. Aggregation and travelling wave dynamics in a two-population model of cancer cell growth and invasion. MATHEMATICAL MEDICINE AND BIOLOGY-A JOURNAL OF THE IMA 2018; 35:541-577. [DOI: 10.1093/imammb/dqx019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 11/14/2017] [Indexed: 12/25/2022]
Affiliation(s)
- Vasiliki Bitsouni
- Division of Mathematics, University of Dundee, Dundee, DD1 4HN, Scotland, UK
| | - Dumitru Trucu
- Division of Mathematics, University of Dundee, Dundee, DD1 4HN, Scotland, UK
| | - Mark A J Chaplain
- School of Mathematics and Statistics, Mathematical Institute (MI), North Haugh
- University of St Andrews, St Andrews, KY16 9SS, Scotland, UK
| | - Raluca Eftimie
- Division of Mathematics, University of Dundee, Dundee, DD1 4HN, Scotland, UK
| |
Collapse
|
16
|
Johansen S, Brenner AK, Bartaula-Brevik S, Reikvam H, Bruserud Ø. The Possible Importance of β3 Integrins for Leukemogenesis and Chemoresistance in Acute Myeloid Leukemia. Int J Mol Sci 2018; 19:ijms19010251. [PMID: 29342970 PMCID: PMC5796198 DOI: 10.3390/ijms19010251] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/20/2017] [Accepted: 01/08/2018] [Indexed: 12/25/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive bone marrow malignancy where the immature leukemia cells communicate with neighboring cells through constitutive cytokine release and through their cell surface adhesion molecules. The primary AML cells express various integrins. These heterodimeric molecules containing an α and a β chain are cell surface molecules that bind extracellular matrix molecules, cell surface molecules and soluble mediators. The β3 integrin (ITGB3) chain can form heterodimers only with the two α chains αIIb and αV. These integrins are among the most promiscuous and bind to a large number of ligands, including extracellular matrix molecules, cell surface molecules and soluble mediators. Recent studies suggest that the two β3 integrins are important for leukemogenesis and chemosensitivity in human AML. Firstly, αIIb and β3 are both important for adhesion of AML cells to vitronectin and fibronectin. Secondly, β3 is important for the development of murine AML and also for the homing and maintenance of the proliferation for xenografted primary human AML cells, and for maintaining a stem cell transcriptional program. These last effects seem to be mediated through Syk kinase. The β3 expression seems to be regulated by HomeboxA9 (HoxA9) and HoxA10, and the increased β3 expression then activates spleen tyrosine kinase (Syk) and thereby contributes to cytokine hypersensitivity and activation of β2 integrins. Finally, high integrin αV/β3 expression is associated with an adverse prognosis in AML and decreased sensitivity to the kinase inhibitor sorafenib; this integrin can also be essential for osteopontin-induced sorafenib resistance in AML. In the present article, we review the experimental and clinical evidence for a role of β3 integrins for leukemogenesis and chemosensitivity in AML.
Collapse
MESH Headings
- Animals
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Disease Models, Animal
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Neoplastic
- Humans
- Integrin beta3/chemistry
- Integrin beta3/genetics
- Integrin beta3/metabolism
- Integrins/chemistry
- Integrins/genetics
- Integrins/metabolism
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/etiology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Ligands
- Multigene Family
- Prognosis
- Protein Binding
- Signal Transduction
Collapse
Affiliation(s)
- Silje Johansen
- Section for Hematology, Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway.
| | - Annette K Brenner
- Section for Hematology, Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway.
- Section for Hematology, Institute of Clinical Science, University of Bergen, 5007 Bergen, Norway.
| | - Sushma Bartaula-Brevik
- Section for Hematology, Institute of Clinical Science, University of Bergen, 5007 Bergen, Norway.
| | - Håkon Reikvam
- Section for Hematology, Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway.
- Section for Hematology, Institute of Clinical Science, University of Bergen, 5007 Bergen, Norway.
| | - Øystein Bruserud
- Section for Hematology, Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway.
- Section for Hematology, Institute of Clinical Science, University of Bergen, 5007 Bergen, Norway.
| |
Collapse
|
17
|
Kim M, Sahu A, Kim GB, Nam GH, Um W, Shin SJ, Jeong YY, Kim IS, Kim K, Kwon IC, Tae G. Comparison of in vivo targeting ability between cRGD and collagen-targeting peptide conjugated nano-carriers for atherosclerosis. J Control Release 2017; 269:337-346. [PMID: 29175140 DOI: 10.1016/j.jconrel.2017.11.033] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 01/04/2023]
Abstract
Atherosclerosis plaque is a major cause of cardiovascular diseases across the globe and a silent killer. There are no physical symptoms of the disease in its early stage and current diagnostic techniques cannot detect the small plaques effectively or safely. Plaques formed in blood vessels can cause serious clinical problems such as impaired blood flow or sudden death, regardless of their size. Thus, detecting early stage of plaques is especially more important to effectively reduce the risk of atherosclerosis. Nanoparticle based delivery systems are recognized as a promising option to fight against this disease, and various targeting ligands are typically used to improve their efficiency. So, the choice of appropriate targeting ligand is a crucial factor for optimal targeting efficiency. cRGD peptide and collagen IV targeting peptide, which binds with the αvβ3 integrin overexpressed in the neovasculature of the plaque and collagen type IV present in the plaque, respectively, are frequently used for the targeting of nanoparticles. However, at present no study has directly compared these two peptides. Therefore, in this study, we have prepared cRGD or collagen IV targeting (Col IV-tg-) peptide conjugated and iron oxide nanoparticle (IONP) loaded Pluronic based nano-carriers for systemic comparison of their targeting ability towards in vivo atherosclerotic plaque in Apolipoprotein E deficient (Apo E-/-) mouse model. Nano-carriers with similar size, surface charge, and IONP loading content but with different targeting ligands were analyzed through in vitro and in vivo experiments. Near infrared fluorescence imaging and magnetic resonance imaging techniques as well as Prussian blue staining were used to compare the accumulation of different ligand conjugated nano-caariers in the aorta of atherosclerotic mice. Our results indicate that cRGD based targeting is more efficient than Col IV-tg-peptide in the early stage of atherosclerosis.
Collapse
Affiliation(s)
- Manse Kim
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Abhishek Sahu
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Gi Beom Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; KU-KIST School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Gi Hoon Nam
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; KU-KIST School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Wooram Um
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - So Jin Shin
- Department of Radiology, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Gwangju 61469, Republic of Korea
| | - Yong Yeon Jeong
- Department of Radiology, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Gwangju 61469, Republic of Korea
| | - In-San Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; KU-KIST School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Kwangmeyung Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; KU-KIST School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Ick Chan Kwon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; KU-KIST School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| |
Collapse
|
18
|
Zajiczek L, Shaw M, Faruqui N, Bella A, Pawar VM, Srinivasan MA, Ryadnov MG. Nano-mechanical single-cell sensing of cell-matrix contacts. NANOSCALE 2016; 8:18105-18112. [PMID: 27734052 DOI: 10.1039/c6nr05667a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Extracellular protein matrices provide a rigidity interface exhibiting nano-mechanical cues that guide cell growth and proliferation. Cells sense such cues using actin-rich filopodia extensions which encourage favourable cell-matrix contacts to recruit more actin-mediated local forces into forming stable focal adhesions. A challenge remains in identifying and measuring these local cellular forces and in establishing empirical relationships between them, cell adhesion and filopodia formation. Here we investigate such relationships using a micromanipulation system designed to operate at the time scale of focal contact dynamics, with the sample frequency of a force probe being 0.1 ms, and to apply and measure forces at nano-to-micro Newton ranges for individual mammalian cells. We explore correlations between cell biomechanics, cell-matrix attachment forces and the spread areas of adhered cells as well as their relative dependence on filopodia formation using synthetic protein matrices with a proven ability to induce enhanced filopodia numbers in adherent cells. This study offers a basis for engineering exploitable cell-matrix contacts in situ at the nanoscale and single-cell levels.
Collapse
Affiliation(s)
- Lydia Zajiczek
- National Physical Laboratory, Hampton Rd, Teddington, Middlesex TW11 0LW, UK.
| | - Michael Shaw
- National Physical Laboratory, Hampton Rd, Teddington, Middlesex TW11 0LW, UK. and UCL TouchLab, Department of Computer Science, University College London, London, WC1E 6BT, UK
| | - Nilofar Faruqui
- National Physical Laboratory, Hampton Rd, Teddington, Middlesex TW11 0LW, UK.
| | - Angelo Bella
- National Physical Laboratory, Hampton Rd, Teddington, Middlesex TW11 0LW, UK.
| | - Vijay M Pawar
- UCL TouchLab, Department of Computer Science, University College London, London, WC1E 6BT, UK
| | - Mandayam A Srinivasan
- UCL TouchLab, Department of Computer Science, University College London, London, WC1E 6BT, UK and MIT TouchLab, Department of Mechanical Engineering and Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Maxim G Ryadnov
- National Physical Laboratory, Hampton Rd, Teddington, Middlesex TW11 0LW, UK.
| |
Collapse
|
19
|
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
|
20
|
Boonstra MC, de Geus SWL, Prevoo HAJM, Hawinkels LJAC, van de Velde CJH, Kuppen PJK, Vahrmeijer AL, Sier CFM. Selecting Targets for Tumor Imaging: An Overview of Cancer-Associated Membrane Proteins. BIOMARKERS IN CANCER 2016; 8:119-133. [PMID: 27721658 PMCID: PMC5040425 DOI: 10.4137/bic.s38542] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/05/2016] [Accepted: 09/06/2016] [Indexed: 12/30/2022]
Abstract
Tumor targeting is a booming business: The global therapeutic monoclonal antibody market accounted for more than $78 billion in 2012 and is expanding exponentially. Tumors can be targeted with an extensive arsenal of monoclonal antibodies, ligand proteins, peptides, RNAs, and small molecules. In addition to therapeutic targeting, some of these compounds can also be applied for tumor visualization before or during surgery, after conjugation with radionuclides and/or near-infrared fluorescent dyes. The majority of these tumor-targeting compounds are directed against cell membrane-bound proteins. Various categories of targetable membrane-bound proteins, such as anchoring proteins, receptors, enzymes, and transporter proteins, exist. The functions and biological characteristics of these proteins determine their location and distribution on the cell membrane, making them more, or less, accessible, and therefore, it is important to understand these features. In this review, we evaluate the characteristics of cancer-associated membrane proteins and discuss their overall usability for cancer targeting, especially focusing on imaging applications.
Collapse
Affiliation(s)
- Martin C Boonstra
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Susanna W L de Geus
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Lukas J A C Hawinkels
- Department of Gastroenterology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands.; Antibodies for Research Applications BV, Gouda, the Netherlands
| | | | - Cornelis F M Sier
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands.; Antibodies for Research Applications BV, Gouda, the Netherlands
| |
Collapse
|
21
|
Herranz-Blanco B, Shahbazi MA, Correia AR, Balasubramanian V, Kohout T, Hirvonen J, Santos HA. pH-Switch Nanoprecipitation of Polymeric Nanoparticles for Multimodal Cancer Targeting and Intracellular Triggered Delivery of Doxorubicin. Adv Healthc Mater 2016; 5:1904-16. [PMID: 27245691 DOI: 10.1002/adhm.201600160] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 04/15/2016] [Indexed: 11/10/2022]
Abstract
Theranostic nanoparticles are emerging as potent tools for noninvasive diagnosis, treatment, and monitoring of solid tumors. Herein, an advanced targeted and multistimuli responsive theranostic platform is presented for the intracellular triggered delivery of doxorubicin. The system consists of a polymeric-drug conjugate solid nanoparticle containing encapsulated superparamagnetic iron oxide nanoparticles (IO@PNP) and decorated with a tumor homing peptide, iRGD. The production of this nanosystem is based on a pH-switch nanoprecipitation method in organic-free solvents, making it ideal for biomedical applications. The nanosystem shows sufficient magnetization saturation for magnetically guided therapy along with reduced cytotoxicity and hemolytic effects. IO@PNP are largely internalized by endothelial and metastatic cancer cells and iRGD decorated IO@PNP moderately enhance their internalization into endothelial cells, while no enhancement is found for the metastatic cancer cells. Poly(ethylene glycol)-block-poly(histidine) with pH-responsive and proton-sponge properties promotes prompt lysosomal escape once the nanoparticles are endocyted. In addition, the polymer-doxorubicin conjugate solid nanoparticles show both intracellular lysosomal escape and efficient translocation of doxorubicin to the nuclei of the cells via cleavage of the amide bond. Overall, IO@PNP-doxorubicin and the iRGD decorated counterpart demonstrate to enhance the toxicity of doxorubicin in cancer cells by improving the intracellular delivery of the drug carried in the IO@PNP.
Collapse
Affiliation(s)
- Bárbara Herranz-Blanco
- Division of Pharmaceutical Chemistry and Technology; Faculty of Pharmacy; University of Helsinki; Viikinkaari 5 E (P. O. Box 56) 00014 Helsinki Finland
| | - Mohammad-Ali Shahbazi
- Division of Pharmaceutical Chemistry and Technology; Faculty of Pharmacy; University of Helsinki; Viikinkaari 5 E (P. O. Box 56) 00014 Helsinki Finland
| | - Alexandra R. Correia
- Division of Pharmaceutical Chemistry and Technology; Faculty of Pharmacy; University of Helsinki; Viikinkaari 5 E (P. O. Box 56) 00014 Helsinki Finland
| | - Vimalkumar Balasubramanian
- Division of Pharmaceutical Chemistry and Technology; Faculty of Pharmacy; University of Helsinki; Viikinkaari 5 E (P. O. Box 56) 00014 Helsinki Finland
| | - Tomáš Kohout
- Department of Physics; University of Helsinki; Gustaf Hällströmin katu 2a (P. O. Box 64) 00560 Helsinki Finland
| | - Jouni Hirvonen
- Division of Pharmaceutical Chemistry and Technology; Faculty of Pharmacy; University of Helsinki; Viikinkaari 5 E (P. O. Box 56) 00014 Helsinki Finland
| | - Hélder A. Santos
- Division of Pharmaceutical Chemistry and Technology; Faculty of Pharmacy; University of Helsinki; Viikinkaari 5 E (P. O. Box 56) 00014 Helsinki Finland
| |
Collapse
|
22
|
Receptor activator of NF-κB ligand induces cell adhesion and integrin α2 expression via NF-κB in head and neck cancers. Sci Rep 2016; 6:23545. [PMID: 27009236 PMCID: PMC4806381 DOI: 10.1038/srep23545] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 03/09/2016] [Indexed: 01/16/2023] Open
Abstract
Cellular interactions with the extracellular matrix play critical roles in tumor progression. We previously reported that receptor activator of NF-κB ligand (RANKL) specifically facilitates head and neck squamous cell carcinoma (HNSCC) progression in vivo. Here, we report a novel role for RANKL in the regulation of cell adhesion. Among the major type I collagen receptors, integrin α2 was significantly upregulated in RANKL-expressing cells, and its knockdown suppressed cell adhesion. The mRNA abundance of integrin α2 positively correlated with that of RANKL in human HNSCC tissues. We also revealed that RANK-NF-κB signaling mediated integrin α2 expression in an autocrine/paracrine manner. Interestingly, the amount of active integrin β1 on the cell surface was increased in RANKL-expressing cells through the upregulation of integrin α2 and endocytosis. Moreover, the RANK-integrin α2 pathway contributed to RANKL-dependent enhanced survival in a collagen gel and inhibited apoptosis in a xenograft model, demonstrating an important role for RANKL-mediated cell adhesion in three-dimensional environments.
Collapse
|
23
|
Zhang D, Qi GB, Zhao YX, Qiao SL, Yang C, Wang H. In Situ Formation of Nanofibers from Purpurin18-Peptide Conjugates and the Assembly Induced Retention Effect in Tumor Sites. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015; 27:6125-30. [PMID: 26350172 DOI: 10.1002/adma.201502598] [Citation(s) in RCA: 258] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/13/2015] [Indexed: 05/23/2023]
Abstract
An assembly-induced retention effect for enhanced tumor photoacoustic (PA) imaging and therapeutics is described. A responsive small-molecule precursor is prepared that simultaneously self-assembles into nanofibers in tumor sites that exhibit an assembly-induced retention effect, which results in an improved PA imaging signal and enhanced therapeutic efficacy. This successful proof-of-concept study paves the way to develop novel supramolecular biomaterials for cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- Di Zhang
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Zhongguancun, 100190, Beijing, China
| | - Guo-Bin Qi
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Zhongguancun, 100190, Beijing, China
| | - Ying-Xi Zhao
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Zhongguancun, 100190, Beijing, China
| | - Sheng-Lin Qiao
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Zhongguancun, 100190, Beijing, China
| | - Chao Yang
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Zhongguancun, 100190, Beijing, China
| | - Hao Wang
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Zhongguancun, 100190, Beijing, China
| |
Collapse
|
24
|
Cockerill M, Rigozzi MK, Terentjev EM. Mechanosensitivity of the 2nd Kind: TGF-β Mechanism of Cell Sensing the Substrate Stiffness. PLoS One 2015; 10:e0139959. [PMID: 26448620 PMCID: PMC4598016 DOI: 10.1371/journal.pone.0139959] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 09/18/2015] [Indexed: 02/01/2023] Open
Abstract
Cells can sense forces applied to them, but also the stiffness of their environment. These are two different phenomena, and here we investigate the mechanosensitivity of the 2nd kind: how the cell can measure an elastic modulus at a single point of adhesion—and how the cell can receive and interpret the chemical signal released from the sensor. Our model uses the example of large latent complex of TGF-β as a sensor. Stochastic theory gives the rate of breaking of latent complex, which initiates the signaling feedback loop after the active TGF-β release and leads to a change of cell phenotype driven by the α-smooth muscle actin. We investigate the dynamic and steady-state behaviors of the model, comparing them with experiments. In particular, we analyse the timescale of approach to the steady state, the stability of the non-linear dynamical system, and how the steady-state concentrations of the key markers vary depending on the elasticity of the substrate. We discover a crossover region for values of substrate elasticity closely corresponding to that of the fibroblast to myofibroblast transition. We suggest that the cell could actively vary the parameters of its dynamic feedback loop to ‘choose’ the position of the transition region and the range of substrate elasticity that it can detect. In this way, the theory offers the unifying mechanism for a variety of phenomena, such as the myofibroblast conversion in fibrosis of wounds and lungs and smooth muscle cell dysfunction in cardiac disease.
Collapse
Affiliation(s)
- Max Cockerill
- Cavendish Laboratory, University of Cambridge, J.J. Thomson Avenue, Cambridge CB3 0HE, United Kingdom
| | - Michelle K. Rigozzi
- Cavendish Laboratory, University of Cambridge, J.J. Thomson Avenue, Cambridge CB3 0HE, United Kingdom
| | - Eugene M. Terentjev
- Cavendish Laboratory, University of Cambridge, J.J. Thomson Avenue, Cambridge CB3 0HE, United Kingdom
- * E-mail:
| |
Collapse
|
25
|
Demirer GS, Okur AC, Kizilel S. Synthesis and design of biologically inspired biocompatible iron oxide nanoparticles for biomedical applications. J Mater Chem B 2015; 3:7831-7849. [PMID: 32262898 DOI: 10.1039/c5tb00931f] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
During the last couple of decades considerable research efforts have been directed towards the synthesis and coating of iron oxide nanoparticles (IONPs) for biomedical applications. To address the current limitations, recent studies have focused on the design of new generation nanoparticle systems whose internalization and targeting capabilities have been improved through surface modifications. This review covers the most recent challenges and advances in the development of IONPs with enhanced quality, and biocompatibility for various applications in biotechnology and medicine.
Collapse
Affiliation(s)
- Gozde S Demirer
- Koc University, Chemical and Biological Engineering, Istanbul 34450, Turkey.
| | | | | |
Collapse
|
26
|
Quantity and accessibility for specific targeting of receptors in tumours. Sci Rep 2014; 4:5232. [PMID: 24912981 PMCID: PMC4050384 DOI: 10.1038/srep05232] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 05/19/2014] [Indexed: 01/03/2023] Open
Abstract
Synaphic (ligand-directed) targeting of drugs is an important potential new approach to drug delivery, particularly in oncology. Considerable success with this approach has been achieved in the treatment of blood-borne cancers, but the advances with solid tumours have been modest. Here, we have studied the number and availability for ligand binding of the receptors for two targeting ligands. The results show that both paucity of total receptors and their poor availability are major bottlenecks in drug targeting. A tumour-penetrating peptide greatly increases the availability of receptors by promoting transport of the drug to the extravascular tumour tissue, but the number of available receptors still remains low, severely limiting the utility of the approach. Our results emphasize the importance of using drugs with high specific activity to avoid exceeding receptor capacity because any excess drug conjugate would lose the targeting advantage. The mathematical models we describe make it possible to focus on those aspects of the targeting mechanism that are most likely to have a substantial effect on the overall efficacy of the targeting.
Collapse
|
27
|
Goswami LN, Ma L, Cai Q, Sarma SJ, Jalisatgi SS, Hawthorne MF. cRGD peptide-conjugated icosahedral closo-B12(2-) core carrying multiple Gd3+-DOTA chelates for α(v)β3 integrin-targeted tumor imaging (MRI). Inorg Chem 2013; 52:1701-9. [PMID: 23391150 PMCID: PMC3593306 DOI: 10.1021/ic302340c] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
A vertex-differentiated icosahedral closo-B(12)(2-) core was utilized to construct a α(v)β(3) integrin receptor-targeted (via cRGD peptide) high payload MRI contrast agent (CA-12) carrying 11 copies of Gd(3+)-DOTA chelates attached to the closo-B(12)(2-) surface via suitable linkers. The resulting polyfunctional MRI contrast agent possessed a higher relaxivity value per-Gd compared to Omniscan, a small molecular contrast agent commonly used in clinical settings. The α(v)β(3) integrin receptor specificity of CA-12 was confirmed via in vitro cellular binding experiments and in vivo MRI of mice bearing human PC-3 prostate cancer xenografts. Integrin α(v)β(3)-positive MDA-MB-231 cells exhibited 300% higher uptake of CA-12 than α(v)β(3)-negative T47D cells. Serial T1-weighted MRI showed superior contrast enhancement of tumors by CA-12 compared to both a nontargeted 12-fold Gd(3+)-DOTA closomer control (CA-7) and Omniscan. Contrast enhancement by CA-12 persisted for 4 h postinjection, and subsequent enhancement of kidney tissue indicated a renal elimination route similar to Omniscan. No toxic effects of CA-12 were apparent in any mice for up to 24 h postinjection. Post-mortem ICP-OES analysis at 24 h detected no residual Gd in any of the tissue samples analyzed.
Collapse
Affiliation(s)
- Lalit N. Goswami
- International Institute of Nano and Molecular Medicine, School of Medicine, University of Missouri, Columbia, Missouri 65211-3450
| | - Lixin Ma
- International Institute of Nano and Molecular Medicine, School of Medicine, University of Missouri, Columbia, Missouri 65211-3450
| | - Quanyu Cai
- International Institute of Nano and Molecular Medicine, School of Medicine, University of Missouri, Columbia, Missouri 65211-3450
| | - Saurav J. Sarma
- International Institute of Nano and Molecular Medicine, School of Medicine, University of Missouri, Columbia, Missouri 65211-3450
| | - Satish S. Jalisatgi
- International Institute of Nano and Molecular Medicine, School of Medicine, University of Missouri, Columbia, Missouri 65211-3450
| | - M. Frederick Hawthorne
- International Institute of Nano and Molecular Medicine, School of Medicine, University of Missouri, Columbia, Missouri 65211-3450
| |
Collapse
|
28
|
Ying CT, Wang J, Lamm RJ, Kamei DT. Mathematical Modeling of Vesicle Drug Delivery Systems 2. ACTA ACUST UNITED AC 2013; 18:46-62. [DOI: 10.1177/2211068212458265] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
29
|
Goubko CA, Basak A, Majumdar S, Jarrell H, Huan Khieu N, Cao X. Comparative analysis of photocaged RGDS peptides for cell patterning. J Biomed Mater Res A 2012; 101:787-96. [DOI: 10.1002/jbm.a.34381] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 06/18/2012] [Indexed: 12/12/2022]
|
30
|
Goodman SL, Grote HJ, Wilm C. Matched rabbit monoclonal antibodies against αv-series integrins reveal a novel αvβ3-LIBS epitope, and permit routine staining of archival paraffin samples of human tumors. Biol Open 2012; 1:329-40. [PMID: 23213423 PMCID: PMC3509452 DOI: 10.1242/bio.2012364] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The relationship between integrin expression and function in pathologies is often contentious as comparisons between human pathological expression and expression in cell lines is difficult. In addition, the expression of even integrins αvβ6 and αvβ8 in tumor cell lines is not comprehensively documented. Here, we describe rabbit monoclonal antibodies (RabMabs) against the extracellular domains of αv integrins that react with both native integrins and formalin fixed, paraffin embedded (FFPE) human tissues. These RabMabs, against αvβ3 (EM22703), αvβ5 (EM09902), αvβ6 (EM05201), αvβ8 (EM13309), and pan-αv (EM01309), recognize individual integrin chains in Western blots and in flow cytometry. EM22703 detected a ligand-induced binding site (LIBS), reporting an epitope enhanced by the binding of an RGD-peptide to αvβ3. αvβ8 was rarely expressed in human tumor specimens, and weakly expressed in non-small-cell lung carcinoma (NSCLC). However, ovarian carcinoma cell lines expressed αvβ8, as did some melanoma cells, whereas U87MG glioma lacked αvβ8 expression. We observed an unexpected strong expression of αvβ6 in tumor samples of invasive ductal breast adenoma, colorectal carcinoma (CRC), and NSCLC. αvβ3 was strongly expressed in some invasive NSCLC cohorts. Interestingly, PC3 prostate cell and human prostate tumors did not express αvβ3. The RabMabs stained plasma membranes in FFPE-immunohistochemistry (IHC) samples of tumor cell lines from lung, ovary, colon, prostate, squamous cell carcinoma of head and neck (SCCHN), breast, and pancreas carcinomas. The RabMabs are unique tools for probing αv integrin biology, and suggest that especially αvβ6 and αvβ8 biologies still have much to reveal.
Collapse
|
31
|
Waite CL, Roth CM. Binding and transport of PAMAM-RGD in a tumor spheroid model: the effect of RGD targeting ligand density. Biotechnol Bioeng 2011; 108:2999-3008. [PMID: 21755497 DOI: 10.1002/bit.23255] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 05/26/2011] [Accepted: 06/20/2011] [Indexed: 11/10/2022]
Abstract
The mechanisms governing the efficient tumor spheroid penetration and transport by poly(amidoamine) (PAMAM) dendrimers displaying varying numbers of cyclic RGD targeting peptides (2, 3, 7, or 10) were evaluated in this work. The cell-free binding affinities and cellular internalization kinetics of PAMAM-RGD conjugates to malignant glioma cells were determined experimentally, and the results were incorporated into a mathematical model to predict the transport of these materials through a multicellular tumor spheroid. The theoretical analysis demonstrated that greater RGD crosslinking may improve transport through tumor spheroids due to their decreased integrin-binding affinity. This study provides evidence that altering the density of tumor-targeting ligands from a drug delivery platform is a feasible way to optimize the tumor-penetration efficiency of an anticancer agent, and provides insight into the physicochemical mechanisms governing the relative effectiveness of these conjugates.
Collapse
Affiliation(s)
- Carolyn L Waite
- Department of Chemical and Biochemical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, New Jersey 08854, USA
| | | |
Collapse
|
32
|
Uchida M, Kosuge H, Terashima M, Willits DA, Liepold LO, Young MJ, McConnell MV, Douglas T. Protein cage nanoparticles bearing the LyP-1 peptide for enhanced imaging of macrophage-rich vascular lesions. ACS NANO 2011; 5:2493-502. [PMID: 21391720 PMCID: PMC3082619 DOI: 10.1021/nn102863y] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Cage-like protein nanoparticles are promising platforms for cell- and tissue-specific targeted delivery of imaging and therapeutic agents. Here, we have successfully modified the 12 nm small heat shock protein from Methanococcus jannaschii (MjHsp) to detect atherosclerotic plaque lesions in a mouse model system. As macrophages are centrally involved in the initiation and progression of atherosclerosis, targeted imaging of macrophages is valuable to assess the biologic status of the blood vessel wall. LyP-1, a nine residue peptide, has been shown to target tumor-associated macrophages. Thus, LyP-1 was genetically incorporated onto the exterior surface of MjHsp, while a fluorescent molecule (Cy5.5) was conjugated on the interior cavity. This bioengineered protein cage, LyP-Hsp, exhibited enhanced affinity to macrophage in vitro. Furthermore, in vivo injection of LyP-Hsp allowed visualization of macrophage-rich murine carotid lesions by in situ and ex vivo fluorescence imaging. These results demonstrate the potential of LyP-1-conjugated protein cages as nanoscale platforms for delivery of imaging agents for the diagnosis of atherosclerosis.
Collapse
Affiliation(s)
- Masaki Uchida
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, 59717
- Department of Plant Sciences, Montana State University, Bozeman, Montana, 59717
| | - Hisanori Kosuge
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Masahiro Terashima
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Deborah A. Willits
- Department of Plant Sciences, Montana State University, Bozeman, Montana, 59717
- Center for Bio-Inspired Nanomaterials (CBIN), Montana State University, Bozeman, Montana, 59717
| | - Lars O. Liepold
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, 59717
- Center for Bio-Inspired Nanomaterials (CBIN), Montana State University, Bozeman, Montana, 59717
| | - Mark J. Young
- Department of Plant Sciences, Montana State University, Bozeman, Montana, 59717
- Center for Bio-Inspired Nanomaterials (CBIN), Montana State University, Bozeman, Montana, 59717
| | - Michael V. McConnell
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Trevor Douglas
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, 59717
- Center for Bio-Inspired Nanomaterials (CBIN), Montana State University, Bozeman, Montana, 59717
- Corresponding author: Trevor Douglas, Professor of Chemistry, Montana State University, Department of Chemistry & Biochemistry, 113 Chemistry and Biochemistry Building, Bozeman, MT 59715, phone (406)994-6566, fax (406) 994-5407,
| |
Collapse
|
33
|
Kessinger CW, Togao O, Khemtong C, Huang G, Takahashi M, Gao J. Investigation of In Vivo Targeting Kinetics of α(v)β(3)-Specific Superparamagnetic Nanoprobes by Time-Resolved MRI. Theranostics 2011; 1:263-73. [PMID: 21562632 PMCID: PMC3092449 DOI: 10.7150/thno/v01p0263] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 03/07/2011] [Indexed: 01/07/2023] Open
Abstract
Nanoparticulate imaging probes have become an increasingly important arsenal in the visualization of molecular markers for early diagnosis and post-therapy assessment of diseases. Surface functionalization of these nanoparticles has led to the development of a variety of targeted nanoprobes for various imaging modalities (e.g. PET, MRI, optical). Despite these advances, detailed understanding of the nanoparticle targeting kinetics, particularly at the early time points immediately after injection, is still lacking. In this study, we report the combination of a T2*-weighted time-resolved-MRI (TR-MRI) method with ultra-sensitive superparamagnetic polymeric micelle (SPPM) nanoprobes to quantify the targeting kinetics of cyclic (RGDfK) (cRGD)-encoded SPPM to angiogenic endothelium in subcutaneous human tumor xenograft models in mice. TR-MRI analyses of the αvβ3-targeted and non-targeted SPPMs allowed for the subtraction of blood volume and extravascular signal components from the cRGD-SPPM data, resulting in a specific measurement of the accumulation kinetics of nanoprobes in lung, breast and brain cancer preclinical models. In all three models, αvβ3-specific accumulation of SPPM nanoprobes was observed in the first 5 mins after intravenous injection (first order rate constants were in the range of 0.22-0.24 min-1). Similar αvβ3-targeting kinetics was observed for cRGD-SPPM nanoprobes in different tumor xenograft models, consistent with the targeting of mouse angiogenic endothelium despite tumor inoculation from different human cancer cell lines. Results from this study offer new opportunities in the quantitative characterization of the targeting kinetics of cancer-specific nanoparticles to their intended biological targets in an intact animal, which provides fundamental insights on molecular recognition processes in vivo for further development of these nanoprobes.
Collapse
|
34
|
Rincón V, Bocanegra R, Rodríguez-Huete A, Rivas G, Mateu MG. Effects of macromolecular crowding on the inhibition of virus assembly and virus-cell receptor recognition. Biophys J 2011; 100:738-746. [PMID: 21281589 PMCID: PMC3030154 DOI: 10.1016/j.bpj.2010.12.3714] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 11/22/2010] [Accepted: 12/15/2010] [Indexed: 10/18/2022] Open
Abstract
Biological fluids contain a very high total concentration of macromolecules that leads to volume exclusion by one molecule to another. Theory and experiment have shown that this condition, termed macromolecular crowding, can have significant effects on molecular recognition. However, the influence of molecular crowding on recognition events involving virus particles, and their inhibition by antiviral compounds, is virtually unexplored. Among these processes, capsid self-assembly during viral morphogenesis and capsid-cell receptor recognition during virus entry into cells are receiving increasing attention as targets for the development of new antiviral drugs. In this study, we have analyzed the effect of macromolecular crowding on the inhibition of these two processes by peptides. Macromolecular crowding led to a significant reduction in the inhibitory activity of: 1), a capsid-binding peptide and a small capsid protein domain that interfere with assembly of the human immunodeficiency virus capsid, and 2), a RGD-containing peptide able to block the interaction between foot-and-mouth disease virus and receptor molecules on the host cell membrane (in this case, the effect was dependent on the conditions used). The results, discussed in the light of macromolecular crowding theory, are relevant for a quantitative understanding of molecular recognition processes during virus infection and its inhibition.
Collapse
Affiliation(s)
- Verónica Rincón
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid), Universidad Autónoma de Madrid, Madrid, Spain
| | - Rebeca Bocanegra
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid), Universidad Autónoma de Madrid, Madrid, Spain
| | - Alicia Rodríguez-Huete
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid), Universidad Autónoma de Madrid, Madrid, Spain
| | - Germán Rivas
- Centro de Investigaciones Biológicas (Consejo Superior de Investigaciones Científicas), Madrid, Spain
| | - Mauricio G Mateu
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid), Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
35
|
Hanaoka K. Development of Responsive Lanthanide-Based Magnetic Resonance Imaging and Luminescent Probes for Biological Applications. Chem Pharm Bull (Tokyo) 2010; 58:1283-94. [DOI: 10.1248/cpb.58.1283] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Kenjiro Hanaoka
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
| |
Collapse
|
36
|
Sugawara K, Yugami A, Kuramitz H. Electrochemical monitoring of binding between wheat germ agglutinin and cellohexose-modified magnetic microbeads. Anal Bioanal Chem 2009; 395:767-72. [DOI: 10.1007/s00216-009-2991-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2009] [Revised: 06/13/2009] [Accepted: 07/17/2009] [Indexed: 01/01/2023]
|
37
|
Park JA, Lee JJ, Jung JC, Yu DY, Oh C, Ha S, Kim TJ, Chang Y. Gd-DOTA Conjugate of RGD as a Potential Tumor-Targeting MRI Contrast Agent. Chembiochem 2008; 9:2811-3. [DOI: 10.1002/cbic.200800529] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
38
|
Multifunctional magnetic nanoparticles for targeted imaging and therapy. Adv Drug Deliv Rev 2008; 60:1241-1251. [PMID: 18508157 DOI: 10.1016/j.addr.2008.03.014] [Citation(s) in RCA: 739] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2007] [Accepted: 03/21/2008] [Indexed: 12/26/2022]
Abstract
Magnetic nanoparticles have become important tools for the imaging of prevalent diseases, such as cancer, atherosclerosis, diabetes, and others. While first generation nanoparticles were fairly nonspecific, newer generations have been targeted to specific cell types and molecular targets via affinity ligands. Commonly, these ligands emerge from phage or small molecule screens, or are based on antibodies or aptamers. Secondary reporters and combined therapeutic molecules have further opened potential clinical applications of these materials. This review summarizes some of the recent biomedical applications of these newer magnetic nanomaterials.
Collapse
|
39
|
Hanaoka K, Lubag AJM, Castillo-Muzquiz A, Kodadek T, Sherry AD. The detection limit of a Gd3+-based T1 agent is substantially reduced when targeted to a protein microdomain. Magn Reson Imaging 2008; 26:608-17. [PMID: 18234462 DOI: 10.1016/j.mri.2007.11.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2007] [Revised: 11/23/2007] [Accepted: 11/24/2007] [Indexed: 10/22/2022]
Abstract
Simple low molecular weight (MW) chelates of Gd(3+) such as those currently used in clinical MRI are considered too insensitive for most molecular imaging applications. Here, we evaluated the detection limit (DL) of a molecularly targeted low MW Gd(3+)-based T(1) agent in a model where the receptor concentration was precisely known. The data demonstrate that receptors clustered together to form a microdomain of high local concentration can be imaged successfully even when the bulk concentration of the receptor is quite low. A GdDO3A-peptide identified by phage display to target the anti-FLAG antibody was synthesized, purified and characterized. T(1-)weighted MR images were compared with the agent bound to antibody in bulk solution and with the agent bound to the antibody localized on agarose beads. Fluorescence competition binding assays show that the agent has a high binding affinity (K(D)=150 nM) for the antibody, while the fully bound relaxivity of the GdDO3A-peptide/anti-FLAG antibody in solution was a relatively modest 17 mM(-1) s(-1). The agent/antibody complex was MR silent at concentrations below approximately 9 microM but was detectable down to 4 microM bulk concentrations when presented to antibody clustered together on the surface of agarose beads. These results provided an estimate of the DLs for other T(1)-based agents with higher fully bound relaxivities or multimeric structures bound to clustered receptor molecules. The results demonstrate that the sensitivity of molecularly targeted contrast agents depends on the local microdomain concentration of the target protein and the molecular relaxivity of the bound complex. A model is presented, which predicts that for a molecularly targeted agent consisting of a single Gd(3+) complex with bound relaxivity of 100 mM(-1) s(-1) or, more reasonably, four tethered Gd(3+) complexes each having a bound relaxivity of 25 mM(-1) s(-1), the DL of a protein microdomain is approximately 690 nM at 9.4 T. These experimental and extrapolated DLs are both well below current literature estimates and suggests that detection of low MW molecularly targeted T(1) agents is not an unrealistic goal.
Collapse
Affiliation(s)
- Kenjiro Hanaoka
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9185, USA
| | | | | | | | | |
Collapse
|
40
|
Main M, Snaith JS, Meloni MM, Jauregui M, Sykes D, Faulkner S, Kenwright AM. Using the Ugi multicomponent condensation reaction to prepare families of chromophore appended azamacrocycles and their complexes. Chem Commun (Camb) 2008:5212-4. [DOI: 10.1039/b810083g] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
41
|
Li ZB, Wu Z, Chen K, Chin FT, Chen X. Click chemistry for (18)F-labeling of RGD peptides and microPET imaging of tumor integrin alphavbeta3 expression. Bioconjug Chem 2007; 18:1987-94. [PMID: 18030991 DOI: 10.1021/bc700226v] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The cell adhesion molecule integrin alpha vbeta 3 plays a key role in tumor angiogenesis and metastasis. A series of (18)F-labeled RGD peptides have been developed for PET of integrin expression based on primary amine reactive prosthetic groups. In this study, we report the use of the Cu(I)-catalyzed Huisgen cycloaddition, also known as a click reaction, to label RGD peptides with (18)F by forming 1,2,3-triazoles. Nucleophilic fluorination of a toluenesulfonic alkyne provided (18)F-alkyne in high yield (nondecay-corrected yield: 65.0 +/- 1.9%, starting from the azeotropically dried (18)F-fluoride), which was then reacted with an RGD azide (nondecay-corrected yield: 52.0 +/- 8.3% within 45 min including HPLC purification). The (18)F-labeled peptide was subjected to microPET studies in murine xenograft models. Murine microPET experiments showed good tumor uptake (2.1 +/- 0.4%ID/g at 1 h postinjection (p.i.)) with rapid renal and hepatic clearance of (18)F-fluoro-PEG-triazoles-RGD 2 ( (18)F-FPTA-RGD2) in a subcutaneous U87MG glioblastoma xenograft model (kidney 2.7 +/- 0.8%ID/g; liver 1.9 +/- 0.4%ID/g at 1 h p.i.). Metabolic stability of the newly synthesized tracer was also analyzed (intact tracer ranging from 75% to 99% at 1 h p.i.). In brief, the new tracer (18)F-FPTA-RGD2 was synthesized with high radiochemical yield and high specific activity. This tracer exhibited good tumor-targeting efficacy and relatively good metabolic stability, as well as favorable in vivo pharmacokinetics. This new (18)F labeling method based on click reaction may also be useful for radiolabeling of other biomolecules with azide groups in high yield.
Collapse
Affiliation(s)
- Zi-Bo Li
- The Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | | | |
Collapse
|