1
|
Duraiyarasu M, Senthil Kumaran S, Mayilmurugan R. Biotin Conjugated Fe(III)-Triscatecholate Complex as Tumour Targeting T 1 MRI Contrast Agent via Second Sphere Water Relaxation. Chemistry 2025; 31:e202403619. [PMID: 39658512 DOI: 10.1002/chem.202403619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/23/2024] [Accepted: 12/10/2024] [Indexed: 12/12/2024]
Abstract
The biotin-conjugated Fe(III) catecholate complex [Fe(BioL)3]3-, Fe(BioL)3 (BioLH2=N-(3,4-dihydroxyphenethyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide) is reported as targeted magnetic resonance imaging (MRI) contrast agents (CAs) to increase the payload for early-stage imaging of tumours. The high spin state and octahedral coordination of the Fe(III) complex are confirmed by EPR spectra and DFT optimized structure, respectively. The overall formation constant (log K) of Fe(BioL)3 is determined as 45, which is higher than the known, more stable complex [Fe(EDTA)]. The complex Fe(BioL)3 exhibited r1 relaxivity of 3.09±0.10 and 1.95±0.09 mM-1 s-1 at 25 °C and 37 °C, respectively, at 1.41 T and pH 7.35 via second-sphere water interactions as due to the absence of inner water coordination. The interaction of Fe(BioL)3 with bovine serum albumin (BSA) and human serum albumin (HSA) displayed enhanced relaxivity of 5.16±0.15 and 10.0±0.19 mM-1 s-1, respectively, at 25 °C and pH 7.3 possibly via lengthening of rotational correlational time. The complex Fe(BioL)3 uptake studies with the avidin, a biotin-binding protein pocket, showed 46 and 37 % enhancements of r1 and r2 relaxivities. Complex Fe(BioL)3 showed 80 % cell viability of the human gastric adenocarcinoma (AGS) cells, and 89 % of iron has been uptaken into cells.
Collapse
Affiliation(s)
- Maheshwaran Duraiyarasu
- Department of Chemistry, & Department of Bioscience & Biomedical Engineering, Indian Institute of Technology Bhilai, Durg, 491002, Chattisgarh, India
- Current Institution, School of Biological and Health Systems Engineering, Arizona State University, Tempe, 85281, Arizona, USA
| | - S Senthil Kumaran
- Department of NMR, All India Institute of Medical Sciences Ansari Nagar, New Delhi, 110 029, India
| | - Ramasamy Mayilmurugan
- Department of Chemistry, & Department of Bioscience & Biomedical Engineering, Indian Institute of Technology Bhilai, Durg, 491002, Chattisgarh, India
| |
Collapse
|
2
|
Cué-Sampedro R, Sánchez-Fernández JA. Functional Post-Synthetic Chemistry of Metal-Organic Cages According to Molecular Architecture and Specific Geometry of Origin. Molecules 2025; 30:462. [PMID: 39942567 PMCID: PMC11820633 DOI: 10.3390/molecules30030462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 02/16/2025] Open
Abstract
Metal-organic cages (MOCs) are discrete supramolecular entities consisting of metal nodes and organic connectors or linkers; MOCs are noted for their high porosity and processability. Chemically, they can be post-synthetically modified (PSM) and new functional groups can be introduced, presenting attractive qualities, and it is expected that their new properties will differ from those of the original compound. This is why they are highly regarded in the fields of biology and chemistry. The present review deals with the current PSM strategies used for MOCs, including covalent, coordination, and noncovalent methods and their structural benefits. The main emphasis of this review is to show to what extent and under what circumstances a MOC can be designed to obtain a tailored geometric architecture. Although sometimes unclear when examining supramolecular systems, particularizing the design of and systematic approaches to the development and characterization of families of MOCs provides new insights into structure-function relationships, which will guide future developments.
Collapse
Affiliation(s)
- Rodrigo Cué-Sampedro
- Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey 64849, Mexico
| | - José Antonio Sánchez-Fernández
- Procesos de Polimerización, Centro de Investigación en Química Aplicada, Blvd. Enrique Reyna No. 140, Saltillo 25294, Mexico
| |
Collapse
|
3
|
Moreno-Alcántar G, Drexler M, Casini A. Assembling a new generation of radiopharmaceuticals with supramolecular theranostics. Nat Rev Chem 2024; 8:893-914. [PMID: 39468298 DOI: 10.1038/s41570-024-00657-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2024] [Indexed: 10/30/2024]
Abstract
Supramolecular chemistry has been used to tackle some of the major challenges in modern science, including cancer therapy and diagnosis. Supramolecular platforms provide synthetic flexibility, rapid generation through self-assembly, facile labelling, unique topologies, tunable reversibility of the enabling noncovalent interactions, and opportunities for host-guest chemistry and mechanical bonding. In this Review, we summarize recent advances in the design and radiopharmaceutical application of discrete self-assembled coordination complexes and mechanically interlocked molecules - namely, metallacages and rotaxanes, respectively - as well as in situ-forming supramolecular aggregates, specifically pinpointing their potential as next-generation radiotheranostic agents. The outlook of such supramolecular constructs for potential applications in the clinic is discussed.
Collapse
Affiliation(s)
- Guillermo Moreno-Alcántar
- Department of Chemistry, School of Natural Sciences, Technical University of Munich, Garching bei München, Germany
| | - Marike Drexler
- Department of Chemistry, School of Natural Sciences, Technical University of Munich, Garching bei München, Germany
| | - Angela Casini
- Department of Chemistry, School of Natural Sciences, Technical University of Munich, Garching bei München, Germany.
- Munich Data Science Institute (MDSI), Technical University of Munich, Garching bei München, Germany.
| |
Collapse
|
4
|
Dissanayake A, Spernyak JA, Morrow JR. An octahedral coordination cage with six Fe(III) centers as a T1 MRI probe. Chem Commun (Camb) 2024; 60:12249-12252. [PMID: 39364604 PMCID: PMC11450543 DOI: 10.1039/d4cc03681f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/23/2024] [Indexed: 10/05/2024]
Abstract
The incorporation of multiple Fe(III) centers bridged by rigid ligands into a coordination cage represents a powerful approach for designing effective MRI contrast agents. In this context, an octahedral coordination cage with six high-spin Fe(III) centers is shown to be water soluble, robust towards dissociation and has effective relaxivity as a T1 MRI probe in solution and in mice.
Collapse
Affiliation(s)
- Aruni Dissanayake
- Department of Chemistry, University at Buffalo, The State University of New York, Amherst, NY 14260, USA.
| | - Joseph A Spernyak
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, USA
| | - Janet R Morrow
- Department of Chemistry, University at Buffalo, The State University of New York, Amherst, NY 14260, USA.
| |
Collapse
|
5
|
Sahoo PR, Spernyak JA, Turowski SG, Morrow JR. Self-Assembled Iron(III) Coordination Cage as an MRI-Active Carrier for a Gold(I) Drug. Bioconjug Chem 2024. [PMID: 39303010 PMCID: PMC11922791 DOI: 10.1021/acs.bioconjchem.4c00391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
A T1 MRI probe based on a self-assembled coordination cage with four iron(III) centers acts as a host for the hydrolysis product of the gold(I) anticancer drug, Au(PEt3)Cl. 1H NMR characterization of the gold complex encapsulated within the diamagnetic Ga(III) analog of the coordination cage is consistent with loss of chloride to give aquated gold complex, most likely [Au(PEt3)(OH2)]+ within the cage. The gold complex undergoes pH-dependent speciation changes in the Ga(III) cage and is released at mildly acidic pH from both the Ga(III) and Fe(III) cages. NMR spectroscopy studies of the encapsulated gold complex in the presence of human serum albumin (HSA) show that the gold complex remains inside of the Ga(III) cage for several hours, resisting release and binding to cysteine residues of HSA. The Fe(III) cage with encapsulated gold complex shows enhanced contrast of the vasculature and uptake into CT26 tumors in BALB/c mice as shown by MRI. The gold complex is solubilized by the iron(III) cage for intravenous injection, whereas the free complex must be injected intraperitoneally. Gold complex accumulates in the tumor for both caged and free complex over 1-48 h as measured by ex-vivo analysis. Encapsulation in the Fe(III) cage modulates the biodistribution of the gold complex in mice in comparison to the free complex, consistent with the function of the cage as a carrier.
Collapse
Affiliation(s)
- Priya Ranjan Sahoo
- Department of Chemistry, University at Buffalo, The State University of New York, Amherst, New York 14260, United States
| | - Joseph A Spernyak
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, United States
| | - Steven G Turowski
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, United States
| | - Janet R Morrow
- Department of Chemistry, University at Buffalo, The State University of New York, Amherst, New York 14260, United States
| |
Collapse
|
6
|
Rinshad V, Aggarwal M, Clegg JK, Mukherjee PS. Harnessing a Pd 4 Water-Soluble Molecular Capsule as a Size-Selective Catalyst for Targeted Oxidation of Alkyl Aromatics. JACS AU 2024; 4:3238-3247. [PMID: 39211591 PMCID: PMC11350579 DOI: 10.1021/jacsau.4c00539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
Molecular hosts with functional cavities can emulate enzymatic behavior through selective encapsulation of substrates, resulting in high chemo-, regio-, and stereoselective product formation. It is still challenging to synthesize enzyme-mimicking hosts that exhibit a narrow substrate scope that relies upon the recognition of substrates based on the molecular size. Herein, we introduce a Pd4 self-assembled water-soluble molecular capsule [M 4 L 2] (MC) that was formed through the self-assembly of a ligand L (4',4‴'-(1,4-phenylene)bis(1',4'-dihydro-[4,2':6',4″-terpyridine]-3',5'-dicarbonitrile)) with the acceptor cis-[(en)Pd(NO3)2] [en = ethane-1,2-diamine] (M). The molecular capsule MC showed size-selective recognition towards xylene isomers. The redox property of MC was explored for efficient and selective oxidation of one of the alkyl groups of m-xylene and p-xylene to their corresponding toluic acids using molecular O2 as an oxidant upon photoirradiation. Employing host-guest chemistry, we demonstrate the homogeneous catalysis of alkyl aromatics to the corresponding monocarboxylic acids in water under mild conditions. Despite homogeneous catalysis, the products were separated from the reaction mixtures by simple filtration/extraction, and the catalyst was reused. The larger analogues of the alkyl aromatics failed to bind within the MC's hydrophobic cavity, resulting in a lower/negligible reaction outcome. The present study represents a facile approach for selective photo-oxidation of xylene isomers to their corresponding toluic acids in an aqueous medium under mild conditions.
Collapse
Affiliation(s)
- Valiyakath
Abdul Rinshad
- Department
of Inorganic and Physical Chemistry, Indian
Institute of Science, Bangalore 560012, India
| | - Medha Aggarwal
- Department
of Inorganic and Physical Chemistry, Indian
Institute of Science, Bangalore 560012, India
| | - Jack K. Clegg
- School
of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Partha Sarathi Mukherjee
- Department
of Inorganic and Physical Chemistry, Indian
Institute of Science, Bangalore 560012, India
| |
Collapse
|
7
|
Yin P, Sun D, Deng Y, Zhu X, Wang Y, Yang J, Feng X. Metal-organic cage as a theranostic nanoplatform for magnetic resonance imaging guided chemodynamic therapy. Theranostics 2024; 14:4861-4873. [PMID: 39239515 PMCID: PMC11373615 DOI: 10.7150/thno.97264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
Rationale: Theranostic nanoplatforms exert a vital role in facilitating concurrent real-time diagnosis and on-demand treatment of diseases, thereby making contributions to the improvement of therapeutic efficacy. Nevertheless, the structural intricacy and the absence of well-defined integration of dual functionality persist as challenges in the development of theranostic nanoplatforms. Methods: We develop an atomically precise theranostic nanoplatform based on metal-organic cage (MOC) to provide magnetic resonance imaging (MRI) guided chemodynamic therapy (CDT) for cancer therapy and assess the theranostic performance both in vitro and in vivo. Through UV-vis spectroscopy, electron paramagnetic resonance (EPR), confocal microscopy, flow cytometry, immunofluorescence staining, and western blotting, the ability of MOC-Mn to generate •OH and the subsequent inhibition of HeLa cells was confirmed. Results: The MOC-Mn composed of manganese and calixarene was successfully synthesized and comprehensively characterized. The catalytic activity of manganese within MOC-Mn facilitated the efficient generation of hydroxyl radicals (•OH) through a Fenton-like reaction, leveraging the high concentrations of hydrogen peroxide in the tumor microenvironment (TME). Additionally, its capacity to prolong the T1 relaxation time and augment the MR signal was observed. The theranostic efficacy was verified via rigorous in vitro and in vivo experiments, indicating that MOC-Mn offered clearer visualization of tumor particulars and substantial suppression of tumor growth. Conclusion: This study showcases a precise MRI-guided CDT theranostic nanoplatform for cancer therapy, thereby promoting the advancement of precise nanomedicine and structure-function research.
Collapse
Affiliation(s)
- Peilin Yin
- School of Pharmacy, China Medical University, Shenyang 110122, China
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- School of Light Industry & Chemical Engineering, Dalian Polytechnic University, Dalian 116034, China
| | - Demei Sun
- School of Pharmacy, China Medical University, Shenyang 110122, China
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yucen Deng
- School of Pharmacy, China Medical University, Shenyang 110122, China
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Youfu Wang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jinghui Yang
- Department of Organ Transplantation, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Xuesong Feng
- School of Pharmacy, China Medical University, Shenyang 110122, China
| |
Collapse
|
8
|
Chakraborty D, Pradhan S, Clegg JK, Mukherjee PS. Mechanically Interlocked Water-Soluble Pd 6 Host for the Selective Separation of Coal Tar-Based Planar Aromatic Molecules. Inorg Chem 2024; 63:14924-14932. [PMID: 39129449 DOI: 10.1021/acs.inorgchem.4c01376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Research on the synthesis of catenated cages has been a growing field of interest in the past few years. While multiple types of catenated cages with different structures have been synthesized, the application of such systems has been much less explored. Specifically, the use of catenated cages in the separation of industrially relevant molecules that are present in coal tar has not been explored before. Herein, we demonstrate the use of a newly synthesized interlocked cage 1 [C184H240N76O48Pd6] (M6L4), formed through the self-assembly of ligand L.HNO3 (tris(4-(1H-imidazole-1-yl)benzylidene)hydrazine-1-carbohydrazonhydrazide) with acceptor cis-[(tmchda)Pd(NO3)2] [tmchda = ±N,N,N',N'-tetramethylcyclohexane-1,2-diamine] (M). The interlocked cage 1 was able to separate the isomers (anthracene and phenanthrene) using a simple solvent extraction technique. Using the same technique, the much more difficult separation of structurally and physiochemically similar compounds acenaphthene and acenaphthylene was performed for the first time with 1 as the host. Other noninterlocked hexanuclear Pd6 cages having a wider cavity proved inefficient for such separation, demonstrating the uniqueness of the interlocked cage 1 for such challenging separation.
Collapse
Affiliation(s)
- Debsena Chakraborty
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India
| | - Sailendra Pradhan
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India
| | - Jack Kay Clegg
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Partha Sarathi Mukherjee
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
9
|
He L, Wang H, Zeng Z, Zhong L, Tang Q, Yu J, Tian J, Liu T, Zhu J. Rigid Fe(III) Chelate with Phosphonate Pendants: A Stable and Effective Extracellular MRI Contrast Agent. J Med Chem 2024; 67:8630-8641. [PMID: 38747630 DOI: 10.1021/acs.jmedchem.3c02338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
A novel Fe(III) complex, Fe-tBPCDTA, was synthesized and explored as a potential contrast agent for MRI. Compared to established agents like Fe-EDTA and Fe-tCDTA, Fe-tBPCDTA exhibited moderate relaxivity (r1 = 1.17 s-1·mmol-1) due to its enhanced second-sphere mechanism. It also displayed improved kinetic inertness, lower cytotoxicity, and enhanced redox stability. In vivo studies demonstrated its function as an extracellular fluid agent, providing tumor contrast comparable to that of Gd-DTPA at a higher dosage. Complete renal clearance occurred within 24 h. These findings suggest Fe-tBPCDTA as a promising candidate for further development as a safe and effective extracellular MRI contrast agent.
Collapse
Affiliation(s)
- Ling He
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
- School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Haiyu Wang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
- School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Zuhua Zeng
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
- School of Pharmacy, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Lei Zhong
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Qingxuan Tang
- Key Laboratory of Synthetic and Self-Assembly Chemistry for Organic Functional Molecules, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Junlai Yu
- Key Laboratory of Synthetic and Self-Assembly Chemistry for Organic Functional Molecules, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Jia Tian
- State Key Laboratory of Organometallic Chemistry and Shanghai Hongkong Joint Laboratory in Chemical Synthesis Key Laboratory of Synthetic and Self-Assembly Chemistry for Organic Functional Molecules, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Tianwei Liu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Jiang Zhu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| |
Collapse
|
10
|
Kras EA, Cineus R, Crawley MR, Morrow JR. Macrocyclic complexes of Fe(III) with mixed hydroxypropyl and phenolate or amide pendants as T 1 MRI probes. Dalton Trans 2024; 53:4154-4164. [PMID: 38318938 PMCID: PMC10897765 DOI: 10.1039/d3dt04013e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/29/2024] [Indexed: 02/07/2024]
Abstract
High-spin Fe(III) complexes of 1,4,7-triazacyclononane (TACN) with mixed oxygen donor pendants including hydroxypropyl, phenolate or amide groups are prepared for study as T1 MRI probes. Complexes with two hydroxypropyl pendants and either amide (Fe(TOAB)) or phenolate (Fe(PTOB)) groups are compared to an analog with three hydroxypropyl groups (Fe(NOHP)), in order to study the effect of the third pendant on the coordination sphere as probed by solution chemistry, relaxivity and structural studies. Solution studies show that Fe(PTOB) has two ionizations with the phenol pendant deprotonating with a pKa of 1.7 and a hydroxypropyl pendent with pKa of 6.3. The X-ray crystal structure of [Fe(PTOB)]Br2 features a six-coordinate complex with two bound hydroxypropyl groups, and a phenolate in a distorted octahedral geometry. The Fe(TOAB) complex has a single deprotonation, assigned to a hydroxypropyl group with a pKa value of 7.0. Both complexes are stabilized as high-spin Fe(III) in solution as shown by their effective magnetic moments and Fe(III)/Fe(II) redox potentials of -390 mV and -780 mV versus NHE at pH 7 and 25 °C for Fe(TOAB) and Fe(PTOB) respectively. Both Fe(PTOB) and Fe(TOAB) are kinetically inert to dissociation under a variety of challenges including phosphate/carbonate buffer, one equivalent of ZnCl2, two equivalents of transferrin or 100 mM HCl, or at basic pH values over 24 h at 37 °C. The r1 relaxivity of Fe(TOAB) at 1.4 T, pH 7.4 and 33 °C is relatively low at 0.6 mM-1 s-1 whereas the r1 relaxivity of Fe(PTOB) is more substantial and shows an increase of 2.5 fold to 2.5 mM-1 s-1 at acidic pH. The increase in relaxivity at acidic pH is attributed to protonation of the phenolate group to provide an additional pathway for proton relaxation.
Collapse
Affiliation(s)
- Elizabeth A Kras
- Department of Chemistry, University at Buffalo, the State University of New York, Amherst, NY 14260, USA.
| | - Roy Cineus
- Department of Chemistry, University at Buffalo, the State University of New York, Amherst, NY 14260, USA.
| | - Matthew R Crawley
- Department of Chemistry, University at Buffalo, the State University of New York, Amherst, NY 14260, USA.
| | - Janet R Morrow
- Department of Chemistry, University at Buffalo, the State University of New York, Amherst, NY 14260, USA.
| |
Collapse
|
11
|
Deiser S, Drexler M, Moreno-Alcántar G, Irl M, Schmidt C, Günther T, Casini A. Synthesis of 177Lu-Labeled, Somatostatin-2 Receptor-Targeted Metalla-Assemblies: Challenges in the Design of Supramolecular Radiotherapeutics. Inorg Chem 2023; 62:20710-20720. [PMID: 37556427 DOI: 10.1021/acs.inorgchem.3c02090] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
Self-assembled supramolecular coordination complexes (SCCs) hold promise for biomedical applications in cancer therapy, although their potential in the field of nuclear medicine is still substantially unexplored. Therefore, in this study an exo-functionalized cationic [Pd2L2]4+ metallacycle (L = 3,5-bis(3-ethynylpyridine)phenyl), targeted to the somatostatin-2 receptor (sst2R) and featuring the DOTA chelator (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) in order to bind the β-- and γ-emitter lutetium-177, was synthesized by self-assembly following ligand synthesis via standard solid-phase peptide synthesis (SPPS). This metallacycle was then characterized by reverse-phase high-performance liquid chromatography (RP-HPLC), electrospray ionization mass spectrometry (ESI-MS), and 1H and 1H-DOSY NMR (DOSY = diffusion-ordered spectroscopy). A procedure for the radiolabeling of the metallacycle with 177Lu was also optimized. The resulting [nat/177Lu]Lu-DOTA-metallacycle, termed [nat/177Lu]Lu-Cy, was evaluated concerning its stability and in vitro properties. The compound was more lipophilic compared to the reference [177Lu]Lu-DOTA-TATE (logPOct/H2O = -0.85 ± 0.10 versus -3.67 ± 0.04, respectively). While [natLu]Lu-Cy revealed low stability in a DMEM/F12 GlutaMax medium, it demonstrated good stability in other aqueous media as well as in DMSO. A high sst2R binding affinity (expressed as IC50) was determined in CHOsst2 cells (Chinese hamster ovary cells that were stably transfected with human sst2R). Moreover, the metallacycle exhibited high human serum albumin binding, as assessed by high-performance affinity chromatography (HPAC), and moderate stability in human serum compared to [177Lu]Lu-DOTA-TATE (TATE = (Tyr3)-octreotate). In order to improve stability, a heteroleptic approach was used to develop a less sterically hindered cage-like SCC that is potentially endowed with host-guest chemistry capability, which has been preliminarily characterized by RP-HPLC and ESI-MS. Overall, our initial results encourage future studies on sst2R-directed SCCs and have led to new insights into the chemistry of ss2R-directed SCCs for radiopharmaceutical applications.
Collapse
Affiliation(s)
- Sandra Deiser
- Chair of Pharmaceutical Radiochemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Walther-Meißner-Str. 3, 85748 Garching b. München, Germany
- Chair of Medicinal and Bioinorganic Chemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching b. München, Germany
| | - Marike Drexler
- Chair of Pharmaceutical Radiochemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Walther-Meißner-Str. 3, 85748 Garching b. München, Germany
- Chair of Medicinal and Bioinorganic Chemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching b. München, Germany
| | - Guillermo Moreno-Alcántar
- Chair of Medicinal and Bioinorganic Chemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching b. München, Germany
| | - Maximilian Irl
- Chair of Pharmaceutical Radiochemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Walther-Meißner-Str. 3, 85748 Garching b. München, Germany
| | - Claudia Schmidt
- Chair of Medicinal and Bioinorganic Chemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching b. München, Germany
| | - Thomas Günther
- Chair of Pharmaceutical Radiochemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Walther-Meißner-Str. 3, 85748 Garching b. München, Germany
| | - Angela Casini
- Chair of Medicinal and Bioinorganic Chemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching b. München, Germany
| |
Collapse
|
12
|
Cineus R, Abozeid SM, Sokolow GE, Spernyak JA, Morrow JR. Fe(III) T1 MRI Probes Containing Phenolate or Hydroxypyridine-Appended Triamine Chelates and a Coordination Site for Bound Water. Inorg Chem 2023; 62:16513-16522. [PMID: 37748050 PMCID: PMC11706235 DOI: 10.1021/acs.inorgchem.3c02344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Fe(III) complexes containing a triamine framework and phenolate or hydroxypyridine donors are characterized and studied as T1 MRI probes. In contrast to most Fe(III) MRI probes of linear chelates reported to date, the ligands reported here are pentadentate to give six-coordinate complexes with a coordination site for inner-sphere water. The crystal structure of the complex containing unsubstituted phenolate donors, Fe(L1)Cl, shows a six-coordinate iron center and contains a chloride ligand that is displaced in water. Two additional derivatives are sufficiently water-soluble for study as MRI probes, including a complex with a hydroxypyridine group, Fe(L2), and a hydroxybenzoic acid group, Fe(L3). The pH potentiometric titrations give protonation constants of 7.2 and 7.5 for Fe(L2) and Fe(L3), respectively, which are assigned to deprotonation of the bound water. Changes in the electronic absorbance spectra of the complexes as a function of pH are consistent with the deprotonation of phenol pendants at acidic pH values. However, the inner-sphere water ligand of Fe(L2) and Fe(L3) does not exchange rapidly on the NMR timescale at pH 6.0 or 7.4, as shown by variable-temperature 17O NMR spectroscopy. The pH-dependent proton relaxivity profiles show a maximum in relaxivity at a near-neutral pH, suggesting that exchange of the protons of the bound water is an important contribution. Competitive binding studies with ethylenediaminetetraacetic acid (EDTA) show effective stability constants for Fe(L2) and Fe(L3) at pH 7.4 with log K values of 21.1 and 20.5, respectively. These two complexes are kinetically inert in carbonate phosphate buffer at 37 °C for several hours but transfer iron to transferrin. Fe(L2) and Fe(L3) show enhanced contrast in T1-weighted imaging analyses in BALB/c mice. These studies show that Fe(L2) clears through mixed renal and hepatobiliary routes, while Fe(L3) has a similar pharmacokinetic clearance profile to a macrocyclic Gd(III) contrast agent.
Collapse
Affiliation(s)
- Roy Cineus
- Department of Chemistry, University at Buffalo, The State University of New York Amherst, New York 14260, United States
| | - Samira M Abozeid
- Department of Chemistry, Faculty of Science, Mansoura University, El-Gomhoria Street, 35516 Mansoura, Egypt
| | - Gregory E Sokolow
- Department of Chemistry, University at Buffalo, The State University of New York Amherst, New York 14260, United States
| | - Joseph A Spernyak
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, United States
| | - Janet R Morrow
- Department of Chemistry, University at Buffalo, The State University of New York Amherst, New York 14260, United States
| |
Collapse
|
13
|
Huang T, Xu X, Cheng C, Wang J, Yang L. Cooperative phototherapy based on bimodal imaging guidance for the treatment of uveal melanoma. J Nanobiotechnology 2023; 21:146. [PMID: 37143039 PMCID: PMC10161622 DOI: 10.1186/s12951-023-01891-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/10/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Uveal melanoma (UM) is adults' most common primary intraocular malignant tumor, prone to metastasis and high mortality. Eyeball enucleation commonly used in the clinic will lead to permanent blindness and mental disorders. Thus, new methods are urgently needed to diagnose and treat UM early to preserve patients' vision. METHODS AND RESULTS Herein, multifunctional nanoparticles (NPs) were synthesized by loading chlorin e6 (Ce6) in poly-lactic-co-glycolic acid (PLGA) NPs and wrapping FeIII-tannic acid (FeIII-TA) on the outside (FeIII-TA/PLGA/Ce6, designated as FTCPNPs). Then, the synergistic photothermal therapy (PTT) and photodynamic therapy (PDT) antitumor effects of FTCPNPs excited by near-infrared (NIR) laser were evaluated. Moreover, we verified the mechanism of synergistic PTT/PDT leading to mitochondrial dysfunction and inducing tumor cell apoptosis. Additionally, FTCPNPs can be used as excellent magnetic resonance (MR)/photoacoustic (PA) imaging contrast agents, enabling imaging-guided cancer treatment. Finally, The NPs have good biological safety. CONCLUSION This noninvasive NIR light-triggered cooperative phototherapy can easily penetrate eye tissue and overcome the disadvantage of limited penetration of phototherapy. Therefore, cooperative phototherapy is expected to be used in fundus tumors. This treatment model is applied to UM for the first time, providing a promising strategy and new idea for integrating the diagnosis and treatment of UM.
Collapse
Affiliation(s)
- Tong Huang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Xinzhi Xu
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing, 400030, P. R. China
| | - Chen Cheng
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Jianxin Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China.
- Department of Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, P. R. China.
| | - Liping Yang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China.
| |
Collapse
|
14
|
Duraiyarasu M, Kumaran SS, Mayilmurugan R. Alkyl Chain Appended Fe(III) Catecholate Complex as a Dual-Modal T1 MRI-NIR Fluorescence Imaging Agent via Second Sphere Water Interactions. ACS Biomater Sci Eng 2023. [PMID: 37141045 DOI: 10.1021/acsbiomaterials.3c00203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The C12-alkyl chain-conjugated Fe(III) catecholate complex [Fe(C12CAT)3]3-, Fe(C12CAT)3 [C12CAT = N-(3,4-dihydroxyphenethyl)dodecanamide], was synthesized and characterized, reported as a dual-modal T1-MRI and an optical imaging probe. The DFT-optimized structure of Fe(C12CAT)3 reveals a distorted octahedral coordination geometry around the high spin Fe(III) center. The formation constant (-log K) of Fe(C12CAT)3 was calculated as 45.4. The complex exhibited r1-relaxivity values of 2.31 ± 0.12 and 1.52 ± 0.06 mM-1 s-1 at 25 and 37 °C, respectively, on 1.41 T at pH 7.3 via second-sphere water interactions. The interaction of Fe(C12CAT)3 with human serum albumin showed concomitant enhancement of r1-relaxivity to 6.44 ± 0.15 mM-1 s-1. The MR phantom images are significantly brighter and directly correlate to the concentration of Fe(C12CAT)3. Adding an external fluorescent marker IR780 dye to Fe(C12CAT)3 leads to the formation of self-assembly by C12-alkyl chains. It resulted in the fluorescence quenching of the dye, and its critical aggregation concentration was calculated as 70 μM. The aggregated matrix of Fe(C12CAT)3 and IR780 dye is spherical, with an average hydrodynamic diameter of 189.5 nm. This self-assembled supramolecular system is found to be non-fluorescent and was "turn-on" under acidic pH via dissociation of aggregates. The r1-relaxivity is found to be unchanged during the matrix aggregation and disaggregation. The probe showed MRI ON and fluorescent OFF under physiological conditions and MRI ON and fluorescent ON under acidic pH. The cell viability experiments showed that the cells are 80% viable at 1 mM probe concentration. Fluorescence experiments and MR phantom images showed that Fe(C12CAT)3 is a potential dual model imaging probe to visualize the acidic pH environment of the cells.
Collapse
Affiliation(s)
- Maheshwaran Duraiyarasu
- Department of Chemistry, and Department of Bioscience & Biomedical Engineering, Indian Institute of Technology Bhilai, Raipur, Chattisgarh 492015, India
| | - S Senthil Kumaran
- Department of NMR, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110 029, India
| | - Ramasamy Mayilmurugan
- Department of Chemistry, and Department of Bioscience & Biomedical Engineering, Indian Institute of Technology Bhilai, Raipur, Chattisgarh 492015, India
| |
Collapse
|
15
|
Huang X, Chen L, Jin J, Kim H, Chen L, Zhang Z, Yu L, Li S, Stang PJ. Host–Guest Encapsulation to Promote the Formation of a Multicomponent Trigonal-Prismatic Metallacage. Inorg Chem 2022; 61:20237-20242. [DOI: 10.1021/acs.inorgchem.2c03701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Affiliation(s)
- Xuechun Huang
- Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Luyi Chen
- Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Jianan Jin
- Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Hyunuk Kim
- Energy Materials and Convergence Research Department, Korea Institute of Energy Research, Daejeon 305-343, Republic of Korea
| | - Luyao Chen
- Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Zibin Zhang
- Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Ling Yu
- Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Shijun Li
- Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Peter J. Stang
- Department of Chemistry, University of Utah, 315 South 1400 East, Room 2020, Salt Lake City, Utah 84112, United States
| |
Collapse
|
16
|
Complementarity and Preorganisation in the Assembly of Heterometallic–Organic Cages via the Metalloligand Approach—Recent Advances. CHEMISTRY 2022. [DOI: 10.3390/chemistry4040095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The design of new metallocage polyhedra towards pre-determined structures can offer both practical as well as intellectual challenges. In this mini-review we discuss a selection of recent examples in which the use of the metalloligand approach has been employed to overcome such challenges. An attractive feature of this approach is its stepwise nature that lends itself to the design and rational synthesis of heterometallic metal–organic cages, with the latter often associated with enhanced functionality.
Collapse
|
17
|
Chakraborty D, Saha R, Clegg JK, Mukherjee PS. Selective separation of planar and non-planar hydrocarbons using an aqueous Pd 6 interlocked cage. Chem Sci 2022; 13:11764-11771. [PMID: 36320911 PMCID: PMC9580621 DOI: 10.1039/d2sc04660a] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 09/13/2022] [Indexed: 11/23/2022] Open
Abstract
Polycyclic aromatic hydrocarbons (PAHs) find multiple applications ranging from fabric dyes to optoelectronic materials. Hydrogenation of PAHs is often employed for their purification or derivatization. However, separation of PAHs from their hydrogenated analogues is challenging because of their similar physical properties. An example of such is the separation of 9,10-dihydroanthracene from phenanthrene/anthracene which requires fractional distillation at high temperature (∼340 °C) to obtain pure anthracene/phenanthrene in coal industry. Herein we demonstrate a new approach for this separation at room temperature using a water-soluble interlocked cage (1) as extracting agent by host-guest chemistry. The cage was obtained by self-assembly of a triimidazole donor L·HNO3 with cis-[(tmeda)Pd(NO3)2] (M) [tmeda = N,N,N',N'-tetramethylethane-1,2-diamine]. 1 has a triply interlocked structure with an inner cavity capable of selectively binding planar aromatic guests.
Collapse
Affiliation(s)
- Debsena Chakraborty
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore 560012 India
| | - Rupak Saha
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore 560012 India
| | - Jack K Clegg
- School of Chemistry and Molecular Biosciences, The University of Queensland St. Lucia Queensland 4072 Australia
| | - Partha Sarathi Mukherjee
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore 560012 India
| |
Collapse
|
18
|
Morrow JR, Raymond JJ, Chowdhury MSI, Sahoo PR. Redox-Responsive MRI Probes Based on First-Row Transition-Metal Complexes. Inorg Chem 2022; 61:14487-14499. [PMID: 36067522 DOI: 10.1021/acs.inorgchem.2c02197] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The presence of multiple oxidation and spin states of first-row transition-metal complexes facilitates the development of switchable MRI probes. Redox-responsive probes capitalize on a change in the magnetic properties of the different oxidation states of the paramagnetic metal ion center upon exposure to biological oxidants and reductants. Transition-metal complexes that are useful for MRI can be categorized according to whether they accelerate water proton relaxation (T1 or T2 agents), induce paramagnetic shifts of 1H or 19F resonances (paraSHIFT agents), or are chemical exchange saturation transfer (CEST) agents. The various oxidation state couples and their properties as MRI probes are summarized with a focus on Co(II)/Co(III) or Fe(II)/Fe(III) complexes as small molecules or as liposomal agents. Solution studies of these MRI probes are reviewed with an emphasis on redox changes upon treatment with oxidants or with enzymes that are physiologically important in inflammation and disease. Finally, we outline the challenges of developing these probes further for in vivo MRI applications.
Collapse
Affiliation(s)
- Janet R Morrow
- Department of Chemistry, University at Buffalo, the State University of New York, Amherst, New York 14260, United States
| | - Jaclyn J Raymond
- Department of Chemistry, University at Buffalo, the State University of New York, Amherst, New York 14260, United States
| | - Md Saiful I Chowdhury
- Department of Chemistry, University at Buffalo, the State University of New York, Amherst, New York 14260, United States
| | - Priya Ranjan Sahoo
- Department of Chemistry, University at Buffalo, the State University of New York, Amherst, New York 14260, United States
| |
Collapse
|
19
|
Chen S, An L, Yang S. Low-Molecular-Weight Fe(III) Complexes for MRI Contrast Agents. Molecules 2022; 27:molecules27144573. [PMID: 35889445 PMCID: PMC9324404 DOI: 10.3390/molecules27144573] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/02/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022] Open
Abstract
Fe(III) complexes have again attracted much attention for application as MRI contrast agents in recent years due to their high thermodynamic stability, low long-term toxicity, and large relaxivity at a higher magnetic field. This mini-review covers the recent progress on low-molecular-weight Fe(III) complexes, which have been considered as one of the promising alternatives to clinically used Gd(III)-based contrast agents. Two kinds of complexes including mononuclear Fe(III) complexes and multinuclear Fe(III) complexes are summarized in sequence, with a specific highlight of the structural relationships between the complexes and their relaxivity and thermodynamic stability. In additional, the future perspectives for the design of low-molecular-weight Fe(III) complexes for MRI contrast agents are suggested.
Collapse
Affiliation(s)
- Shangjun Chen
- Key Laboratory of Resource Chemistry of Ministry of Education, Shanghai Key Laboratory of Rare Earth Functional Materials, and Department of Chemistry, Shanghai Normal University, Shanghai 200234, China;
| | - Lu An
- Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors, Shanghai Normal University, Shanghai 200234, China;
| | - Shiping Yang
- Key Laboratory of Resource Chemistry of Ministry of Education, Shanghai Key Laboratory of Rare Earth Functional Materials, and Department of Chemistry, Shanghai Normal University, Shanghai 200234, China;
- Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors, Shanghai Normal University, Shanghai 200234, China;
- Correspondence:
| |
Collapse
|
20
|
Kras EA, Snyder EM, Sokolow GE, Morrow JR. Distinct Coordination Chemistry of Fe(III)-Based MRI Probes. Acc Chem Res 2022; 55:1435-1444. [PMID: 35482819 DOI: 10.1021/acs.accounts.2c00102] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
ConspectusContrast agents are used in approximately 40% of all magnetic resonance imaging (MRI) procedures to improve the quality of the images based on the distribution and dynamic clearance of the agent. To date, all clinically approved contrast agents are Gd(III) coordination complexes that serve to shorten the longitudinal (T1) and transverse (T2) proton relaxation times of water. Recent interest in replacing Gd with biologically relevant metal ions such as Mn or Fe has led to increased interest in the aqueous coordination chemistry of their complexes. In this Account, we focus on high-spin Fe(III) complexes that have been recently reported as MRI contrast agents or probes in our laboratory.The highly Lewis acidic Fe(III) center has distinct coordination chemistry in aqueous solutions, facilitating alternative strategies in the design of MRI probes. To illustrate this, we describe different classes of Fe(III) MRI probes with a focus on macrocyclic complexes and multinuclear complexes such as self-assembled metal organic polyhedra (MOP). Our initial efforts focused on macrocyclic complexes of Fe(III) in order to tune spin and oxidation states with the goal of stabilizing high-spin Fe(III) in reducing biological environments. Our probes feature six-coordinate Fe(III) complexes of 1,4,7-triazacyclononane with hydroxypropyl, phosphonate, or carboxylate pendant groups to produce Fe(III) complexes that shorten proton T1 times predominantly from second-sphere or outer-sphere interactions at neutral pH. Analogues with pentadentate macrocyclic ligands have an inner-sphere water that does not exchange rapidly on the NMR time scale, yet these complexes are effective relaxation agents. Fe(III) macrocyclic complexes in this class can be modified to modulate their biodistribution and pharmacokinetic clearance in mice. The goal of these studies is for the Fe(III) agents to clear as extracellular fluid agents and produce profiles similar to those of Gd agents. Finally, studies of multimeric Fe(III) complexes are of interest to produce probes that give large proton relaxivity. In this approach the two Fe(III) centers are connected through aryl linkers as demonstrated for several macrocyclic complexes. Even more tightly connected Fe(III) centers are produced in a Fe(III) self-assembled cage with relaxivity of 21 mM-1 s-1 at 4.7 T, 37 °C in the presence of serum albumin to which it is tightly bound. This cage enhances contrast of the vasculature as a blood pool agent and accumulates in tumors. Finally, we present our perspectives on the further development of Fe(III) complexes for various applications in MRI.
Collapse
Affiliation(s)
- Elizabeth A. Kras
- Department of Chemistry, University at Buffalo, the State University of New York, Amherst, New York 14260, United States
| | - Eric M. Snyder
- Department of Chemistry, University at Buffalo, the State University of New York, Amherst, New York 14260, United States
| | - Gregory E. Sokolow
- Department of Chemistry, University at Buffalo, the State University of New York, Amherst, New York 14260, United States
| | - Janet R. Morrow
- Department of Chemistry, University at Buffalo, the State University of New York, Amherst, New York 14260, United States
| |
Collapse
|
21
|
Jayapaul J, Komulainen S, Zhivonitko VV, Mareš J, Giri C, Rissanen K, Lantto P, Telkki VV, Schröder L. Hyper-CEST NMR of metal organic polyhedral cages reveals hidden diastereomers with diverse guest exchange kinetics. Nat Commun 2022; 13:1708. [PMID: 35361759 PMCID: PMC8971460 DOI: 10.1038/s41467-022-29249-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 03/03/2022] [Indexed: 01/04/2023] Open
Abstract
Guest capture and release are important properties of self-assembling nanostructures. Over time, a significant fraction of guests might engage in short-lived states with different symmetry and stereoselectivity and transit frequently between multiple environments, thereby escaping common spectroscopy techniques. Here, we investigate the cavity of an iron-based metal organic polyhedron (Fe-MOP) using spin-hyperpolarized 129Xe Chemical Exchange Saturation Transfer (hyper-CEST) NMR. We report strong signals unknown from previous studies that persist under different perturbations. On-the-fly delivery of hyperpolarized gas yields CEST signatures that reflect different Xe exchange kinetics from multiple environments. Dilute pools with ~ 104-fold lower spin numbers than reported for directly detected hyperpolarized nuclei are readily detected due to efficient guest turnover. The system is further probed by instantaneous and medium timescale perturbations. Computational modeling indicates that these signals originate likely from Xe bound to three Fe-MOP diastereomers (T, C3, S4). The symmetry thus induces steric effects with aperture size changes that tunes selective spin manipulation as it is employed in CEST MRI agents and, potentially, impacts other processes occurring on the millisecond time scale.
Collapse
Affiliation(s)
- Jabadurai Jayapaul
- Molecular Imaging, Department of Structural Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany
- Division of Translational Molecular Imaging, Deutsches Krebsforschungszentrum (DKFZ), 69120, Heidelberg, Germany
| | | | | | - Jiří Mareš
- NMR Research Unit, University of Oulu, 90014, Oulu, Finland
- Research Unit of Medical Imaging, Physics and Technology (MIPT), University of Oulu, 90014, Oulu, Finland
| | - Chandan Giri
- University of Jyvaskyla, Department of Chemistry, 40014, Jyväskylä, Finland
| | - Kari Rissanen
- University of Jyvaskyla, Department of Chemistry, 40014, Jyväskylä, Finland
| | - Perttu Lantto
- NMR Research Unit, University of Oulu, 90014, Oulu, Finland.
| | | | - Leif Schröder
- Molecular Imaging, Department of Structural Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany.
- Division of Translational Molecular Imaging, Deutsches Krebsforschungszentrum (DKFZ), 69120, Heidelberg, Germany.
| |
Collapse
|