1
|
Zeng T, Wu Q, Liu Y, Qi Q, Shen W, Gu W, Zhang Y, Xiong W, Xie Z, Qi X, Tian T, Zhou X. Unraveling the Cleavage Reaction of Hydroxylamines with Cyclopropenones Considering Biocompatibility. J Am Chem Soc 2024; 146:35077-35089. [PMID: 39660762 DOI: 10.1021/jacs.4c09757] [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: 12/12/2024]
Abstract
We develop a latent biocompatible cleavage reaction involving the hitherto unexplored interaction between hydroxylamines and cyclopropenones. Our study addresses the regioselectivity challenges commonly observed in asymmetric cyclopropenone transformations, substantiated by variations in substrate, Density Functional Theory calculations, and in situ NMR analysis. This reaction is characterized by high efficiency, broad substrate scope, stability, latent biocompatibility, and mild reaction conditions. Significantly, it facilitates fluorescence activation and functions as a controlled release mechanism for prodrugs, showing great promise in biological assays. Our success in achieving the controlled release of nitrogen mustard in HeLa cells underscores its potential application in cellular contexts. Additionally, we introduce a simple and highly efficient method for synthesizing α, β-substituted pentenolides, applicable to a variety of substrates. Moreover, we extend this cleavage reaction to the CRISPR-Cas9 system, achieving precise, on-demand regulation of guide RNA activity. The introduction of this cleavage reaction offers a promising tool for biochemical research and biotechnological applications.
Collapse
Affiliation(s)
- Tianying Zeng
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Quan Wu
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Yongjie Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Qianqian Qi
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Wei Shen
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Wei Gu
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Yuanyuan Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Wei Xiong
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Zhongpao Xie
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Xiaotian Qi
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Tian Tian
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Xiang Zhou
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| |
Collapse
|
2
|
Sancho-Albero M, Sebastian V, Perez-Lopez AM, Martin-Duque P, Unciti-Broceta A, Santamaria J. Extracellular Vesicles-Mediated Bio-Orthogonal Catalysis in Growing Tumors. Cells 2024; 13:691. [PMID: 38667306 PMCID: PMC11048864 DOI: 10.3390/cells13080691] [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: 03/11/2024] [Revised: 04/10/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Several studies have reported the successful use of bio-orthogonal catalyst nanoparticles (NPs) for cancer therapy. However, the delivery of the catalysts to the target tissues in vivo remains an unsolved challenge. The combination of catalytic NPs with extracellular vesicles (EVs) has been proposed as a promising approach to improve the delivery of therapeutic nanomaterials to the desired organs. In this study, we have developed a nanoscale bio-hybrid vector using a CO-mediated reduction at low temperature to generate ultrathin catalytic Pd nanosheets (PdNSs) as catalysts directly inside cancer-derived EVs. We have also compared their biodistribution with that of PEGylated PdNSs delivered by the EPR effect. Our results indicate that the accumulation of PdNSs in the tumour tissue was significantly higher when they were administered within the EVs compared to the PEGylated PdNSs. Conversely, the amount of Pd found in non-target organs (i.e., liver) was lowered. Once the Pd-based catalytic EVs were accumulated in the tumours, they enabled the activation of a paclitaxel prodrug demonstrating their ability to carry out bio-orthogonal uncaging chemistries in vivo for cancer therapy.
Collapse
Affiliation(s)
- Maria Sancho-Albero
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Avda. San Juan Bosco, 13, 50009 Zaragoza, Spain; (V.S.); (J.S.)
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Campus Rio Ebro, Edificio I+D, C/Poeta Mariano Esquillor, s/n, 50018 Zaragoza, Spain
- Networking Research Center in Biomaterials, Bioengineering and Nanomedicine (CIBERBBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Chemical and Enviromental Engineering, University of Zaragoza, Campus Rio Ebro, C/María de Luna, 3, 50018 Zaragoza, Spain
| | - Victor Sebastian
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Avda. San Juan Bosco, 13, 50009 Zaragoza, Spain; (V.S.); (J.S.)
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Campus Rio Ebro, Edificio I+D, C/Poeta Mariano Esquillor, s/n, 50018 Zaragoza, Spain
- Networking Research Center in Biomaterials, Bioengineering and Nanomedicine (CIBERBBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Chemical and Enviromental Engineering, University of Zaragoza, Campus Rio Ebro, C/María de Luna, 3, 50018 Zaragoza, Spain
| | - Ana M. Perez-Lopez
- Edinburgh Cancer Research, CRUK Scotland Centre, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh EH4 2XR, UK; (A.M.P.-L.); (A.U.-B.)
| | - Pilar Martin-Duque
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Avda. San Juan Bosco, 13, 50009 Zaragoza, Spain; (V.S.); (J.S.)
- Networking Research Center in Biomaterials, Bioengineering and Nanomedicine (CIBERBBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Instituto de Salud Carlos III, 28222 Madrid, Spain
| | - Asier Unciti-Broceta
- Edinburgh Cancer Research, CRUK Scotland Centre, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh EH4 2XR, UK; (A.M.P.-L.); (A.U.-B.)
| | - Jesus Santamaria
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Avda. San Juan Bosco, 13, 50009 Zaragoza, Spain; (V.S.); (J.S.)
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Campus Rio Ebro, Edificio I+D, C/Poeta Mariano Esquillor, s/n, 50018 Zaragoza, Spain
- Networking Research Center in Biomaterials, Bioengineering and Nanomedicine (CIBERBBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Chemical and Enviromental Engineering, University of Zaragoza, Campus Rio Ebro, C/María de Luna, 3, 50018 Zaragoza, Spain
| |
Collapse
|
3
|
Dal Forno GM, Latocheski E, Navo CD, Albuquerque BL, St John AL, Avenier F, Jiménez-Osés G, Domingos JB. Interplay of chloride levels and palladium(ii)-catalyzed O-deallenylation bioorthogonal uncaging reactions. Chem Sci 2024; 15:4458-4465. [PMID: 38516072 PMCID: PMC10952092 DOI: 10.1039/d3sc06408e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/15/2024] [Indexed: 03/23/2024] Open
Abstract
The palladium-mediated uncaging reaction of allene substrates remains a promising yet often overlooked strategy in the realm of bioorthogonal chemistry. This method exhibits high kinetic rates, rivaling those of the widely employed allylic and propargylic protecting groups. In this study, we investigate into the mechanistic aspects of the C-O bond-cleavage deallenylation reaction, examining how chloride levels influence the kinetics when triggered by Pd(ii) complexes. Focusing on the deallenylation of 1,2-allenyl protected 4-methylumbelliferone promoted by Allyl2Pd2Cl2, our findings reveal that reaction rates are higher in environments with lower chloride concentrations, mirroring intracellular conditions, compared to elevated chloride concentrations typical of extracellular conditions. Through kinetic and spectroscopic experiments, combined with DFT calculations, we uncover a detailed mechanism that identifies AllylPd(H2O)2 as the predominant active species. These insights provide the basis for the design of π-allylpalladium catalysts suited for selective uncaging within specific cellular environments, potentially enhancing targeted therapeutic applications.
Collapse
Affiliation(s)
- Gean M Dal Forno
- Laboratory of Biomimetic Catalysis (LaCBio), Department of Chemistry, Federal University of Santa Catarina (UFSC) Campus Trindade Florianópolis 88040-900 SC Brazil
| | - Eloah Latocheski
- Laboratory of Biomimetic Catalysis (LaCBio), Department of Chemistry, Federal University of Santa Catarina (UFSC) Campus Trindade Florianópolis 88040-900 SC Brazil
| | - Claudio D Navo
- Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance (BRTA) Bizkaia Technology Park, Building 800, Derio 48160 Spain
| | - Brunno L Albuquerque
- Laboratory of Biomimetic Catalysis (LaCBio), Department of Chemistry, Federal University of Santa Catarina (UFSC) Campus Trindade Florianópolis 88040-900 SC Brazil
| | - Albert L St John
- Laboratory of Biomimetic Catalysis (LaCBio), Department of Chemistry, Federal University of Santa Catarina (UFSC) Campus Trindade Florianópolis 88040-900 SC Brazil
| | - Frédéric Avenier
- Institut de Chimie Moléculaire et des Matériaux d'Orsay (UMR 8182), Université Paris Saclay 9140 Orsay Cedex France
| | - Gonzalo Jiménez-Osés
- Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance (BRTA) Bizkaia Technology Park, Building 800, Derio 48160 Spain
- Ikerbasque, Basque Foundation for Science 48013 Bilbao Spain
| | - Josiel B Domingos
- Laboratory of Biomimetic Catalysis (LaCBio), Department of Chemistry, Federal University of Santa Catarina (UFSC) Campus Trindade Florianópolis 88040-900 SC Brazil
| |
Collapse
|
4
|
Braun J, Ortega-Liebana MC, Unciti-Broceta A, Sieber SA. A Pd-labile fluoroquinolone prodrug efficiently prevents biofilm formation on coated surfaces. Org Biomol Chem 2024; 22:1998-2002. [PMID: 38375536 DOI: 10.1039/d4ob00014e] [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: 02/21/2024]
Abstract
Surface-adhered bacteria on implants represent a major challenge for antibiotic treatment. We introduce hydrogel-coated surfaces loaded with tailored Pd-nanosheets which catalyze the release of antibiotics from inactive prodrugs. Masked and antibiotically inactive fluoroquinolone analogs were efficiently activated at the surface and prevented the formation of Staphylococcus aureus biofilms.
Collapse
Affiliation(s)
- Josef Braun
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Center for Functional Protein Assemblies (CPA), Ernst-Otto-Fischer Strasse 8, 85748 Garching bei München, Germany.
| | - M Carmen Ortega-Liebana
- Edinburgh Cancer Research, Institute of Genetics and Cancer, University of Edinburgh, EH4 2XR Edinburgh, UK
- CRUK Scotland Centre, UK
- Department of Medicinal & Organic Chemistry and Unit of Excellence in Chemistry Applied to Biomedicine and the Environment, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain
- GENYO, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
| | - Asier Unciti-Broceta
- Edinburgh Cancer Research, Institute of Genetics and Cancer, University of Edinburgh, EH4 2XR Edinburgh, UK
- CRUK Scotland Centre, UK
| | - Stephan A Sieber
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Center for Functional Protein Assemblies (CPA), Ernst-Otto-Fischer Strasse 8, 85748 Garching bei München, Germany.
| |
Collapse
|
5
|
Huang R, Hirschbiegel CM, Lehot V, Liu L, Cicek YA, Rotello VM. Modular Fabrication of Bioorthogonal Nanozymes for Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2300943. [PMID: 37042795 PMCID: PMC11234510 DOI: 10.1002/adma.202300943] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/21/2023] [Indexed: 06/19/2023]
Abstract
The incorporation of transition metal catalysts (TMCs) into nanoscaffolds generates nanocatalysts that replicate key aspects of enzymatic behavior. The TMCs can access bioorthogonal chemistry unavailable to living systems. These bioorthogonal nanozymes can be employed as in situ "factories" for generating bioactive molecules where needed. The generation of effective bioorthogonal nanozymes requires co-engineering of the TMC and the nanometric scaffold. This review presents an overview of recent advances in the field of bioorthogonal nanozymes, focusing on modular design aspects of both nanomaterial and catalyst and how they synergistically work together for in situ uncaging of imaging and therapeutic agents.
Collapse
Affiliation(s)
- Rui Huang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Cristina-Maria Hirschbiegel
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Victor Lehot
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Liang Liu
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Yagiz Anil Cicek
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| |
Collapse
|
6
|
Tan Y, Pierrard F, Frédérick R, Riant O. Enhancing Tsuji-Trost deallylation in living cells with an internal-nucleophile coumarin-based probe. RSC Adv 2024; 14:5492-5498. [PMID: 38352674 PMCID: PMC10862660 DOI: 10.1039/d3ra08938j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 01/31/2024] [Indexed: 02/16/2024] Open
Abstract
In recent years, bioorthogonal uncaging reactions have been developed to proceed efficiently under physiological conditions. However, limited progress has been made in the development of protecting groups combining stability under physiological settings with the ability to be quickly removed via bioorthogonal catalysis. Herein, we present a new water-soluble coumarin-derived probe bearing an internal nucleophilic group capable of promoting Tsuji-Trost deallylation under palladium catalysis. This probe can be cleaved by a bioorthogonal palladium complex at a faster rate than the traditional probe, namely N-Alloc-7-amino-4-methylcoumarin. As the deallylation process proved to be efficient in mammalian cells, we envision that this probe may find applications in chemical biology, bioengineering, and medicine.
Collapse
Affiliation(s)
- Yonghua Tan
- Institute of Condensed Matter and Nanosciences (IMCN), Université catholique de Louvain Louvain-la-Neuve 1348 Belgium
- Louvain Drug Research Institute (LDRI), Université catholique de Louvain Brussels B-1200 Belgium
| | - François Pierrard
- Institute of Condensed Matter and Nanosciences (IMCN), Université catholique de Louvain Louvain-la-Neuve 1348 Belgium
- Louvain Drug Research Institute (LDRI), Université catholique de Louvain Brussels B-1200 Belgium
| | - Raphaël Frédérick
- Louvain Drug Research Institute (LDRI), Université catholique de Louvain Brussels B-1200 Belgium
| | - Olivier Riant
- Institute of Condensed Matter and Nanosciences (IMCN), Université catholique de Louvain Louvain-la-Neuve 1348 Belgium
| |
Collapse
|
7
|
Nabawy A, Gupta A, Jiang M, Hirschbiegel CM, Fedeli S, Chattopadhyay AN, Park J, Zhang X, Liu L, Rotello VM. Biodegradable nanoemulsion-based bioorthogonal nanocatalysts for intracellular generation of anticancer therapeutics. NANOSCALE 2023; 15:13595-13602. [PMID: 37554065 PMCID: PMC10528015 DOI: 10.1039/d3nr01801f] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
Bioorthogonal catalysis mediated by transition metal catalysts (TMCs) provides controlled in situ activation of prodrugs through chemical reactions that do not interfere with cellular bioprocesses. The direct use of 'naked' TMCs in biological environments can have issues of solubility, deactivation, and toxicity. Here, we demonstrate the design and application of a biodegradable nanoemulsion-based scaffold stabilized by a cationic polymer that encapsulates a palladium-based TMC, generating bioorthogonal nanocatalyst "polyzymes". These nanocatalysts enhance the stability and catalytic activity of the TMCs while maintaining excellent mammalian cell biocompatibility. The therapeutic potential of these nanocatalysts was demonstrated through efficient activation of a non-toxic prodrug into an active chemotherapeutic drug, leading to efficient killing of cancer cells.
Collapse
Affiliation(s)
- Ahmed Nabawy
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA.
| | - Aarohi Gupta
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA.
| | - Mingdi Jiang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA.
| | - Cristina-Maria Hirschbiegel
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA.
| | - Stefano Fedeli
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA.
| | - Aritra Nath Chattopadhyay
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA.
| | - Jungmi Park
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA.
| | - Xianzhi Zhang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA.
| | - Liang Liu
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA.
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA.
| |
Collapse
|
8
|
Hirschbiegel CM, Zhang X, Huang R, Cicek YA, Fedeli S, Rotello VM. Inorganic nanoparticles as scaffolds for bioorthogonal catalysts. Adv Drug Deliv Rev 2023; 195:114730. [PMID: 36791809 PMCID: PMC10170407 DOI: 10.1016/j.addr.2023.114730] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023]
Abstract
Bioorthogonal transition metal catalysts (TMCs) transform therapeutically inactive molecules (pro-drugs) into active drug compounds. Inorganic nanoscaffolds protect and solubilize catalysts while offering a flexible design space for decoration with targeting elements and stimuli-responsive activity. These "drug factories" can activate pro-drugs in situ, localizing treatment to the disease site and minimizing off-target effects. Inorganic nanoscaffolds provide structurally diverse scaffolds for encapsulating TMCs. This ability to define the catalyst environment can be employed to enhance the stability and selectivity of the TMC, providing access to enzyme-like bioorthogonal processes. The use of inorganic nanomaterials as scaffolds TMCs and the use of these bioorthogonal nanozymes in vitro and in vivo applications will be discussed in this review.
Collapse
Affiliation(s)
| | - Xianzhi Zhang
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St, Amherst, MA 01003, USA
| | - Rui Huang
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St, Amherst, MA 01003, USA
| | - Yagiz Anil Cicek
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St, Amherst, MA 01003, USA
| | - Stefano Fedeli
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St, Amherst, MA 01003, USA
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St, Amherst, MA 01003, USA.
| |
Collapse
|
9
|
Sathyan A, Deng L, Loman T, Palmans AR. Bio-orthogonal catalysis in complex media: Consequences of using polymeric scaffold materials on catalyst stability and activity. Catal Today 2023. [DOI: 10.1016/j.cattod.2023.114116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
|
10
|
Sousa-Castillo A, Mariño-López A, Puértolas B, Correa-Duarte MA. Nanostructured Heterogeneous Catalysts for Bioorthogonal Reactions. Angew Chem Int Ed Engl 2023; 62:e202215427. [PMID: 36479797 DOI: 10.1002/anie.202215427] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/05/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022]
Abstract
Bioorthogonal chemistry has inspired a new subarea of chemistry providing a powerful tool to perform novel biocompatible chemospecific reactions in living systems. Following the premise that they do not interfere with biological functions, bioorthogonal reactions are increasingly applied in biomedical research, particularly with respect to genetic encoding systems, fluorogenic reactions for bioimaging, and cancer therapy. This Minireview compiles recent advances in the use of heterogeneous catalysts for bioorthogonal reactions. The synthetic strategies of Pd-, Au-, and Cu-based materials, their applicability in the activation of caged fluorophores and prodrugs, and the possibilities of using external stimuli to release therapeutic substances at a specific location in a diseased tissue are discussed. Finally, we highlight frontiers in the field, identifying challenges, and propose directions for future development in this emerging field.
Collapse
|
11
|
Ji X, Zhong Z. External stimuli-responsive gasotransmitter prodrugs: Chemistry and spatiotemporal release. J Control Release 2022; 351:81-101. [PMID: 36116579 DOI: 10.1016/j.jconrel.2022.09.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/28/2022]
Abstract
Gasotransmitters like nitric oxide, carbon monoxide, and hydrogen sulfide with unique pleiotropic pharmacological effects in mammals are an emerging therapeutic modality for different human diseases including cancer, infection, ischemia-reperfusion injuries, and inflammation; however, their clinical translation is hampered by the lack of a reliable delivery form, which delivers such gasotransmitters to the action site with precisely controlled dosage. The external stimuli-responsive prodrug strategy has shown tremendous potential in developing gasotransmitter prodrugs, which affords precise temporospatial control and better dose control compared with endogenous stimuli-sensitive prodrugs. The promising external stimuli employed for gasotransmitter activation range from photo, ultrasound, and bioorthogonal click chemistry to exogenous enzymes. Herein, we highlight the recent development of external stimuli-mediated decaging chemistry for the temporospatial delivery of gasotransmitters including nitric oxide, carbon monoxide, hydrogen sulfide and sulfur dioxide, and discuss the pros and cons of different designs.
Collapse
Affiliation(s)
- Xingyue Ji
- College of Pharmaceutical Sciences, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China.
| | - Zhiyuan Zhong
- College of Pharmaceutical Sciences, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
12
|
Huang R, Hirschbiegel CM, Zhang X, Gupta A, Fedeli S, Xu Y, Rotello VM. Engineered Polymer-Supported Biorthogonal Nanocatalysts Using Flash Nanoprecipitation. ACS APPLIED MATERIALS & INTERFACES 2022; 14:31594-31600. [PMID: 35802797 DOI: 10.1021/acsami.2c04496] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Transition-metal catalysts (TMCs) effect bioorthogonal transformations that enable the generation of therapeutic agents in situ, minimizing off-target effects. The encapsulation of insoluble TMCs into polymeric nanoparticles to generate "polyzymes" has vastly expanded their applicability in biological environments by enhancing catalyst solubility and stability. However, commonly used precipitation approaches provide limited encapsulation efficiency in polyzyme fabrication and result in a low catalytic activity. Herein, we report the creation of polyzymes with increased catalyst loading and optimized turnover efficiency using flash nanoprecipitation (FNP). Polyzymes with controlled size and catalyst loading were fabricated by tuning the process conditions of FNP. The biological applicability of polyzymes was demonstrated by efficiently transforming a non-toxic prodrug into the active drug within cancer cells.
Collapse
Affiliation(s)
- Rui Huang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Cristina-Maria Hirschbiegel
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Xianzhi Zhang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Aarohi Gupta
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Stefano Fedeli
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Yisheng Xu
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237 P. R. China
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| |
Collapse
|
13
|
Zhang X, Lin S, Huang R, Gupta A, Fedeli S, Cao-Milán R, Luther DC, Liu Y, Jiang M, Li G, Rondon B, Wei H, Rotello VM. Degradable ZnS-Supported Bioorthogonal Nanozymes with Enhanced Catalytic Activity for Intracellular Activation of Therapeutics. J Am Chem Soc 2022; 144:12893-12900. [PMID: 35786910 DOI: 10.1021/jacs.2c04571] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Bioorthogonal catalysis using transition-metal catalysts (TMCs) provides a toolkit for the in situ generation of imaging and therapeutic agents in biological environments. Integrating TMCs with nanomaterials mimics key properties of natural enzymes, providing bioorthogonal "nanozymes". ZnS nanoparticles provide a platform for bioorthogonal nanozymes using ruthenium catalysts embedded in self-assembled monolayers on the particle surface. These nanozymes uncage allylated profluorophores and prodrugs. The ZnS core combines the non-toxicity and degradability with the enhancement of Ru catalysis through the release of thiolate surface ligands that accelerate the rate-determining step in the Ru-mediated deallylation catalytic cycle. The maximum rate of reaction (Vmax) increases ∼2.5-fold as compared to the non-degradable gold nanoparticle analogue. The therapeutic potential of these bioorthogonal nanozymes is demonstrated by activating a chemotherapy drug from an inactive prodrug with efficient killing of cancer cells.
Collapse
Affiliation(s)
- Xianzhi Zhang
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Shichao Lin
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States.,Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, People's Republic of China.,State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, Jiangsu 210023, People's Republic of China
| | - Rui Huang
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Aarohi Gupta
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Stefano Fedeli
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Roberto Cao-Milán
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
| | - David C Luther
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Yuanchang Liu
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Mingdi Jiang
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Gengtan Li
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Brayan Rondon
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Hui Wei
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, People's Republic of China.,State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, Jiangsu 210023, People's Republic of China
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
| |
Collapse
|
14
|
Liu Y, Lai KL, Vong K. Transition Metal Scaffolds Used To Bring New‐to‐Nature Reactions into Biological Systems. Eur J Inorg Chem 2022. [DOI: 10.1002/ejic.202200215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Yifei Liu
- Department of Chemistry The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon Hong Kong China
| | - Ka Lun Lai
- Department of Chemistry The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon Hong Kong China
| | - Kenward Vong
- Department of Chemistry The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon Hong Kong China
| |
Collapse
|
15
|
Adam C, Bray TL, Pérez-López AM, Tan EH, Rubio-Ruiz B, Baillache DJ, Houston DR, Salji MJ, Leung HY, Unciti-Broceta A. A 5-FU Precursor Designed to Evade Anabolic and Catabolic Drug Pathways and Activated by Pd Chemistry In Vitro and In Vivo. J Med Chem 2022; 65:552-561. [PMID: 34979089 DOI: 10.1021/acs.jmedchem.1c01733] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
5-Fluorouracil (5-FU) is an antineoplastic antimetabolite that is widely administered to cancer patients by bolus injection, especially to those suffering from colorectal and pancreatic cancer. Because of its suboptimal route of administration and dose-limiting toxicities, diverse 5-FU prodrugs have been developed to confer oral bioavailability and increase the safety profile of 5-FU chemotherapy regimens. Our contribution to this goal is presented herein with the development of a novel palladium-activated prodrug designed to evade the metabolic machinery responsible for 5-FU anabolic activation and catabolic processing. The new prodrug is completely innocuous to cells and highly resistant to metabolization by primary hepatocytes and liver S9 fractions (the main metabolic route for 5-FU degradation), whereas it is rapidly converted into 5-FU in the presence of a palladium (Pd) source. In vivo pharmokinetic analysis shows the prodrug is rapidly and completely absorbed after oral administration and exhibits a longer half-life than 5-FU. In vivo efficacy studies in a xenograft colon cancer model served to prove, for the first time, that orally administered prodrugs can be locally converted to active drugs by intratumorally inserted Pd implants.
Collapse
Affiliation(s)
- Catherine Adam
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, EH4 2XU Edinburgh, U.K
| | - Thomas L Bray
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, EH4 2XU Edinburgh, U.K
| | - Ana M Pérez-López
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, EH4 2XU Edinburgh, U.K
| | - Ee Hong Tan
- Institute of Cancer Sciences, University of Glasgow, Bearsden, Glasgow G61 1QH, U.K.,Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, Glasgow G61 1BD, U.K
| | - Belén Rubio-Ruiz
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, EH4 2XU Edinburgh, U.K
| | - Daniel J Baillache
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, EH4 2XU Edinburgh, U.K
| | - Douglas R Houston
- Institute of Quantitative Biology, Biochemistry and Biotechnology, University of Edinburgh, Edinburgh EH9 3BF, U.K
| | - Mark J Salji
- Institute of Cancer Sciences, University of Glasgow, Bearsden, Glasgow G61 1QH, U.K.,Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, Glasgow G61 1BD, U.K
| | - Hing Y Leung
- Institute of Cancer Sciences, University of Glasgow, Bearsden, Glasgow G61 1QH, U.K.,Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, Glasgow G61 1BD, U.K
| | - Asier Unciti-Broceta
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, EH4 2XU Edinburgh, U.K
| |
Collapse
|
16
|
Nasibullin I, Smirnov I, Ahmadi P, Vong K, Kurbangalieva A, Tanaka K. Synthetic prodrug design enables biocatalytic activation in mice to elicit tumor growth suppression. Nat Commun 2022; 13:39. [PMID: 35013295 PMCID: PMC8748823 DOI: 10.1038/s41467-021-27804-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/03/2021] [Indexed: 11/25/2022] Open
Abstract
Considering the intrinsic toxicities of transition metals, their incorporation into drug therapies must operate at minimal amounts while ensuring adequate catalytic activity within complex biological systems. As a way to address this issue, this study investigates the design of synthetic prodrugs that are not only tuned to be harmless, but can be robustly transformed in vivo to reach therapeutically relevant levels. To accomplish this, retrosynthetic prodrug design highlights the potential of naphthylcombretastatin-based prodrugs, which form highly active cytostatic agents via sequential ring-closing metathesis and aromatization. Structural adjustments will also be done to improve aspects related to catalytic reactivity, intrinsic bioactivity, and hydrolytic stability. The developed prodrug therapy is found to possess excellent anticancer activities in cell-based assays. Furthermore, in vivo activation by intravenously administered glycosylated artificial metalloenzymes can also induce significant reduction of implanted tumor growth in mice.
Collapse
Affiliation(s)
- Igor Nasibullin
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Ivan Smirnov
- Biofunctional Chemistry Laboratory, A. Butlerov Institute of Chemistry, Kazan Federal University, 18 Kremlyovskaya street, Kazan, 420008, Russia
| | - Peni Ahmadi
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Kenward Vong
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Almira Kurbangalieva
- Biofunctional Chemistry Laboratory, A. Butlerov Institute of Chemistry, Kazan Federal University, 18 Kremlyovskaya street, Kazan, 420008, Russia
| | - Katsunori Tanaka
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan.
- Biofunctional Chemistry Laboratory, A. Butlerov Institute of Chemistry, Kazan Federal University, 18 Kremlyovskaya street, Kazan, 420008, Russia.
- GlycoTargeting Research Laboratory, RIKEN Baton Zone Program, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan.
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 O-okayama, Meguro-ku, Tokyo, 152-8552, Japan.
| |
Collapse
|
17
|
Konč J, Sabatino V, Jiménez‐Moreno E, Latocheski E, Pérez LR, Day J, Domingos JB, Bernardes GJL. Controlled In‐Cell Generation of Active Palladium(0) Species for Bioorthogonal Decaging. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202113519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Juraj Konč
- Yusuf Hamied Department of Chemistry University of Cambridge Lensfield Road CB2 1EW Cambridge UK
| | - Valerio Sabatino
- Yusuf Hamied Department of Chemistry University of Cambridge Lensfield Road CB2 1EW Cambridge UK
| | - Ester Jiménez‐Moreno
- Yusuf Hamied Department of Chemistry University of Cambridge Lensfield Road CB2 1EW Cambridge UK
| | - Eloah Latocheski
- LaCBio—Laboratory of Biomimetic Catalysis Department of Chemistry Federal University of Santa Catarina—UFSC Campus Trindade SC 88040–900 Florianópolis Brazil
| | - Laura Rodríguez Pérez
- Yusuf Hamied Department of Chemistry University of Cambridge Lensfield Road CB2 1EW Cambridge UK
| | - Jason Day
- Department of Earth Sciences University of Cambridge Downing Street CB2 3EQ Cambridge UK
| | - Josiel B. Domingos
- LaCBio—Laboratory of Biomimetic Catalysis Department of Chemistry Federal University of Santa Catarina—UFSC Campus Trindade SC 88040–900 Florianópolis Brazil
| | - Gonçalo J. L. Bernardes
- Yusuf Hamied Department of Chemistry University of Cambridge Lensfield Road CB2 1EW Cambridge UK
- Instituto de Medicina Molecular João Lobo Antunes Faculdade de Medicina Universidade de Lisboa Avenida Professor Egas Moniz 1649-028 Lisboa Portugal
| |
Collapse
|
18
|
Tevet S, Wagle SS, Slor G, Amir RJ. Tuning the Reactivity of Micellar Nanoreactors by Precise Adjustments of the Amphiphile and Substrate Hydrophobicity. Macromolecules 2021; 54:11419-11426. [PMID: 34987270 PMCID: PMC8717824 DOI: 10.1021/acs.macromol.1c01755] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/02/2021] [Indexed: 01/12/2023]
Abstract
Polymeric assemblies, such as micelles, are gaining increasing attention due to their ability to serve as nanoreactors for the execution of organic reactions in aqueous media. The ability to conduct organic transformations, which have been traditionally limited to organic media, in water is essential for the further development of important fields ranging from green catalysis to bioorthogonal chemistry. Considering the recent progress that has been made to expand the range of organometallic reactions conducted using nanoreactors, we aimed to gain a deeper understanding of the roles of the hydrophobicity of both the core of micellar nanoreactors and the substrates on the reaction rates in water. Toward this goal, we designed a set of five metal-loaded micelles composed of polyethylene glycol-dendron amphiphiles and studied their ability to serve as nanoreactors for a palladium-mediated depropargylation reaction of four substrates with different log P values. Using dendrons as the hydrophobic block, we could precisely tune the lipophilicity of the nanoreactors, which allowed us to reveal linear correlations between the rate constants and the hydrophobicity of the amphiphiles (estimated by the dendron's cLog P). While exponential dependence was obtained for the lipophilicity of the substrates, a similar degree of rate acceleration was observed due to the increase in the hydrophobicity of the amphiphiles regardless of the effect of the substrate's log P. Our results demonstrate that while increasing the hydrophobicity of the substrates may be used to accelerate reaction rates, tuning the hydrophobicity of the micellar nanoreactors can serve as a vital tool for further optimization of the reactivity and selectivity of nanoreactors.
Collapse
Affiliation(s)
- Shahar Tevet
- Department
of Organic Chemistry, School of Chemistry, Faculty of Exact Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
- Tel-Aviv
University Center for Nanoscience and Nanotechnology, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Shreyas S. Wagle
- Department
of Organic Chemistry, School of Chemistry, Faculty of Exact Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
- Tel-Aviv
University Center for Nanoscience and Nanotechnology, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Gadi Slor
- Department
of Organic Chemistry, School of Chemistry, Faculty of Exact Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
- Tel-Aviv
University Center for Nanoscience and Nanotechnology, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Roey J. Amir
- Department
of Organic Chemistry, School of Chemistry, Faculty of Exact Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
- Tel-Aviv
University Center for Nanoscience and Nanotechnology, Tel-Aviv University, Tel-Aviv 6997801, Israel
- Blavatnik
Center for Drug Discovery, Tel-Aviv University, Tel-Aviv 6997801, Israel
- ADAMA
Center for Novel Delivery Systems in Crop Protection, Tel-Aviv University, Tel-Aviv 6997801, Israel
- The
Center for Physics and Chemistry of Living Systems, Tel-Aviv University, Tel-Aviv 6997801, Israel
| |
Collapse
|
19
|
Fedeli S, Im J, Gopalakrishnan S, Elia JL, Gupta A, Kim D, Rotello VM. Nanomaterial-based bioorthogonal nanozymes for biological applications. Chem Soc Rev 2021; 50:13467-13480. [PMID: 34787131 PMCID: PMC8862209 DOI: 10.1039/d0cs00659a] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bioorthogonal transformations are chemical reactions that use pathways which biological processes do not access. Bioorthogonal chemistry provides new approaches for imaging and therapeutic strategies, as well as tools for fundamental biology. Bioorthogonal catalysis enables the development of bioorthogonal "factories" for on-demand and in situ generation of drugs and imaging tools. Transition metal catalysts (TMCs) are widely employed as bioorthogonal catalysts due to their high efficiency and versatility. The direct application of TMCs in living systems is challenging, however, due to their limited solubility, instability in biological media and toxicity. Incorporation of TMCs into nanomaterial scaffolds can be used to enhance aqueous solubility, improve long-term stability in biological environment and minimize cytotoxicity. These nanomaterial platforms can be engineered for biomedical applications, increasing cellular uptake, directing biodistribution, and enabling active targeting. This review summarizes strategies for incorporating TMCs into nanomaterial scaffolds, demonstrating the potential and challenges of moving bioorthogonal nanocatalysts and nanozymes toward the clinic.
Collapse
Affiliation(s)
- Stefano Fedeli
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Jungkyun Im
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States,Department of Chemical Engineering, 22 Soonchunhyangro, Soonchunhyang University, Asan, 31538, Republic of Korea,Department of Electronic Materials and Devices Engineering, 22 Soonchunhyangro, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Sanjana Gopalakrishnan
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - James L. Elia
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Aarohi Gupta
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Dongkap Kim
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States,Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea,Department of Chemistry, Hanyang University, Seoul, 04763, Republic of Korea
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| |
Collapse
|
20
|
Konč J, Sabatino V, Jiménez-Moreno E, Latocheski E, Pérez LR, Day J, Domingos JB, Bernardes GJL. Controlled In-Cell Generation of Active Palladium(0) Species for Bioorthogonal Decaging. Angew Chem Int Ed Engl 2021; 61:e202113519. [PMID: 34739737 DOI: 10.1002/anie.202113519] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Indexed: 11/07/2022]
Abstract
Owing to their bioorthogonality, transition metals have become very popular in the development of biocompatible bond-cleavage reactions. However, many approaches require design and synthesis of complex ligands or formulation of nanoparticles which often perform poorly in living cells. This work reports on a method for the generation of an active palladium species that triggers bond-cleaving reactions inside living cells. We utilized the water-soluble Na2PdCl4 as a simple source of Pd(II) which can be intracellularly reduced by sodium ascorbate to the active Pd(0) species. Once generated, Pd(0) triggers the cleavage of allyl ether and carbamate caging groups leading to the release of biologically active molecules. These findings do not only expand the toolbox of available bioorthogonal dissociative reactions but also provide an additional strategy for controlling the reactivity of Pd species involved in Pd-mediated bioorthogonal reactions.
Collapse
Affiliation(s)
- Juraj Konč
- University of Cambridge, Chemistry, UNITED KINGDOM
| | | | | | | | | | - Jason Day
- University of Cambridge, Earth Sciences, UNITED KINGDOM
| | | | - Gonçalo J L Bernardes
- University of Cambridge, Yusuf Hamied Department of Chemistry, Lensfield Road, CB21EW, Cambridge, UNITED KINGDOM
| |
Collapse
|
21
|
In vivo organic synthesis by metal catalysts. Bioorg Med Chem 2021; 46:116353. [PMID: 34419820 DOI: 10.1016/j.bmc.2021.116353] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/19/2021] [Accepted: 08/02/2021] [Indexed: 11/21/2022]
Abstract
The metal-catalyzed reactions have given various chemical modifications that could not be achieved through basic organic chemistry reactions. In the past decade, many metal-mediated catalytic systems have carried out different transformations in cellulo, such as decaging of fluorophores, drug release, and protein conjugation. However, translating abiotic metal catalysts for organic synthesis in vivo, including bacteria, zebrafish, or mice, could encounter numerous challenges regarding their biocompatibility, stability, and reactivity in the complicated biological environment. In this review, we categorize and summarize the relevant advances in this research field by emphasizing the system's framework, the design of each transformation, and the mode of action. These studies disclose the massive potential of the emerging field and the significant applications in synthetic biology.
Collapse
|
22
|
Wang W, Zhang X, Huang R, Hirschbiegel CM, Wang H, Ding Y, Rotello VM. In situ activation of therapeutics through bioorthogonal catalysis. Adv Drug Deliv Rev 2021; 176:113893. [PMID: 34333074 PMCID: PMC8440397 DOI: 10.1016/j.addr.2021.113893] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 07/01/2021] [Accepted: 07/20/2021] [Indexed: 12/29/2022]
Abstract
Bioorthogonal chemistry refers to any chemical reactions that can occur inside of living systems without interfering with native biochemical processes, which has become a promising strategy for modulating biological processes. The development of synthetic metal-based catalysts to perform bioorthogonal reactions has significantly expanded the toolkit of bioorthogonal chemistry for medicinal chemistry and synthetic biology. A wide range of homogeneous and heterogeneous transition metal catalysts (TMCs) have been reported, mediating different transformations such as cycloaddition reactions, as well as bond forming and cleaving reactions. However, the direct application of 'naked' TMCs in complex biological media poses numerous challenges, including poor water solubility, toxicity and catalyst deactivation. Incorporating TMCs into nanomaterials to create bioorthogonal nanocatalysts can solubilize and stabilize catalyst molecules, with the decoration of the nanocatalysts used to provide spatiotemporal control of catalysis. This review presents an overview of the advances in the creation of bioorthogonal nanocatalysts, highlighting different choice of nano-scaffolds, and the therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Wenjie Wang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Xianzhi Zhang
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Rui Huang
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | | | - Huaisong Wang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Ya Ding
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China.
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA.
| |
Collapse
|
23
|
Wang J, Wang X, Fan X, Chen PR. Unleashing the Power of Bond Cleavage Chemistry in Living Systems. ACS CENTRAL SCIENCE 2021; 7:929-943. [PMID: 34235254 PMCID: PMC8227596 DOI: 10.1021/acscentsci.1c00124] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Indexed: 05/02/2023]
Abstract
Bioorthogonal cleavage chemistry has been rapidly emerging as a powerful tool for manipulation and gain-of-function studies of biomolecules in living systems. While the initial bond formation-centered bioorthogonal reactions have been widely adopted for labeling, tracing, and capturing biomolecules, the newly developed bond cleavage-enabled bioorthogonal reactions have opened new possibilities for rescuing small molecules as well as biomacromolecules in living systems, allowing multidimensional controls over biological processes in vitro and in vivo. In this Outlook, we first summarized the development and applications of bioorthogonal cleavage reactions (BCRs) that restore the functions of chemical structures as well as more complex networks, including the liberation of prodrugs, release of bioconjugates, and in situ reactivation of intracellular proteins. As we embarked on this fruitful progress, we outlined the unmet scientific needs and future directions along this exciting avenue. We believe that the potential of BCRs will be further unleashed when combined with other frontier technologies, such as genetic code expansion and proximity-enabled chemical labeling.
Collapse
Affiliation(s)
- Jie Wang
- Beijing
National Laboratory for Molecular Sciences, Synthetic and Functional
Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular
Engineering of Ministry of Education, College of Chemistry and Molecular
Engineering, Peking University, Beijing 100871, China
- Department
of Chemistry, Southern University of Science
and Technology, Shenzhen 518055, China
| | - Xin Wang
- Beijing
National Laboratory for Molecular Sciences, Synthetic and Functional
Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular
Engineering of Ministry of Education, College of Chemistry and Molecular
Engineering, Peking University, Beijing 100871, China
| | - Xinyuan Fan
- Beijing
National Laboratory for Molecular Sciences, Synthetic and Functional
Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular
Engineering of Ministry of Education, College of Chemistry and Molecular
Engineering, Peking University, Beijing 100871, China
| | - Peng R. Chen
- Beijing
National Laboratory for Molecular Sciences, Synthetic and Functional
Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular
Engineering of Ministry of Education, College of Chemistry and Molecular
Engineering, Peking University, Beijing 100871, China
- Peking−Tsinghua
Center for Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
24
|
Mancuso F, Rahm M, Dzijak R, Mertlíková-Kaiserová H, Vrabel M. Transition-Metal-Mediated versus Tetrazine-Triggered Bioorthogonal Release Reactions: Direct Comparison and Combinations Thereof. Chempluschem 2021; 85:1669-1675. [PMID: 32757364 DOI: 10.1002/cplu.202000477] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/16/2020] [Indexed: 01/14/2023]
Abstract
Bioorthogonal cleavage reactions are gaining popularity in chemically inducible prodrug activation and in the control of biomolecular functions. Despite similar applications, these reactions were developed and optimized on different substrates and under different experimental conditions. Reported herein is a side-by-side comparison of palladium-, ruthenium- and tetrazine-triggered release reactions, which aims at comparing the reaction kinetics, efficiency and overall advantages and limitations of the methods. In addition, we disclose the possibility of mutual combination of the cleavage reactions. Finally, we compare the efficiency of the bioorthogonal deprotections in cellular experiments, which revealed that among the three methods investigated, the palladium- and the tetrazine-promoted reaction can be used for efficient prodrug activation, but only the tetrazine-triggered reactions proceed efficiently inside cells.
Collapse
Affiliation(s)
- Francesca Mancuso
- University of Messina, Department of Chemical, Biological Pharmaceutical and Environmental Sciences (CHIBIOFARAM), Viale Palatucci 13, I-98168, Messina, Italy
| | - Michal Rahm
- Institute of Organic Chemistry, and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 166 10, Prague, Czech Republic
| | - Rastislav Dzijak
- Institute of Organic Chemistry, and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 166 10, Prague, Czech Republic
| | - Helena Mertlíková-Kaiserová
- Institute of Organic Chemistry, and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 166 10, Prague, Czech Republic
| | - Milan Vrabel
- Institute of Organic Chemistry, and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 166 10, Prague, Czech Republic
| |
Collapse
|
25
|
Oladipo AO, Unuofin JO, Iku SII, Nkambule TTI, Mamba BB, Msagati TAM. Bimetallic Au@Pd nanodendrite system incorporating multimodal intracellular imaging for improved doxorubicin antitumor efficiency. Int J Pharm 2021; 602:120661. [PMID: 33933638 DOI: 10.1016/j.ijpharm.2021.120661] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 11/19/2022]
Abstract
The sufficient accumulation of drugs is crucial for efficient treatment in a complex tumor microenvironment. Drug delivery systems (DDS) with high surface area and selective cytotoxicity present a novel approach to mitigate insufficient drug loading for improved therapeutic response. Herein, a doxorubicin-conjugated bimetallic gold-core palladium-shell nanocarrier with multiple dense arrays of branches (Au@PdNDs.PEG/DOX) was characterized and its efficacy against breast adenocarcinoma (MCF-7) and lung adenocarcinoma (A549) cells were evaluated. Enhanced darkfield and hyperspectral imaging (HSI) microscopy were used to study the intracellular uptake and accumulation of the DOX-loaded nanodendrites A fascinating data from a 3D-CytoViva fluorescence imaging technique provided information about the dynamics of localization and distribution of the nanocarrier. In vitro cytotoxicity assays indicated that Au@PdNDs.PEG/DOX inhibited the proliferative effects of MCF-7 cells at equivalent IC50 dosage compared to DOX alone. The nanocarrier triggered higher induction of apoptosis proved by a time-dependent phosphatidylserine V release, cell cycle arrest, and flow cytometry analysis. Moreover, the cell cycle phase proportion increase suggests that the enhanced apoptotic effect induced by Au@PdNDs.PEG/DOX was via a G2/M phase arrest. Thus, this study demonstrated the potential of dendritic nanoparticles to improve DOX therapeutic efficiency and plasmonic-mediated intracellular imaging as a suitable theranostic platform for deployment in nanomedicine.
Collapse
Affiliation(s)
- Adewale O Oladipo
- Institute for Nanotechnology and Water Sustainability (iNanoWS), College of Science, Engineering and Technology, University of South Africa, Science Park Florida, Johannesburg 1710, South Africa.
| | - Jeremiah O Unuofin
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Science Park Florida, Johannesburg 1710, South Africa
| | - Solange I I Iku
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Science Park Florida, Johannesburg 1710, South Africa
| | - Thabo T I Nkambule
- Institute for Nanotechnology and Water Sustainability (iNanoWS), College of Science, Engineering and Technology, University of South Africa, Science Park Florida, Johannesburg 1710, South Africa
| | - Bhekie B Mamba
- Institute for Nanotechnology and Water Sustainability (iNanoWS), College of Science, Engineering and Technology, University of South Africa, Science Park Florida, Johannesburg 1710, South Africa
| | - Titus A M Msagati
- Institute for Nanotechnology and Water Sustainability (iNanoWS), College of Science, Engineering and Technology, University of South Africa, Science Park Florida, Johannesburg 1710, South Africa.
| |
Collapse
|
26
|
Wu J, Sun T, Yang C, Lv T, Bi Y, Xu Y, Ling Y, Zhao J, Cong R, Zhang Y, Wang J, Wen H, Jiang H, Li F, Huang Z. Tetrazine-mediated bioorthogonal removal of 3-isocyanopropyl groups enables the controlled release of nitric oxide in vivo. Biomater Sci 2021; 9:1816-1825. [PMID: 33458722 DOI: 10.1039/d0bm01841d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Bond cleavage bioorthogonal chemistry has been widely employed to restore or activate proteins or prodrugs. Nitric oxide (NO), as a free radical molecule, has joined the clinical arena of cancer therapy, since high levels of NO could produce a cancer cell growth inhibitory effect. However, the spatiotemporal controlled release of NO remains a great challenge, and bioorthogonal chemistry may open a new window. Herein, we described a class of O2-3-isocyanopropyl diazeniumdiolates 3a-f as new bioorthogonal NO precursors, which can be effectively uncaged via tetrazine-mediated bond cleavage reactions to liberate NO and acrolein in living cancer cells, exhibiting potent antiproliferative activity. Furthermore, 3a and tetrazine BTZ were respectively encapsulated into two liposomes. It was found that simultaneous administrations of the two liposomes could specifically release large amounts of NO in the implanted cancer cells in zebrafish, thus generating potent tumor suppression activity in vivo. Our findings indicate that the TZ-labile NO precursors could serve to expand the NO-based smart therapeutics and the scope of bioorthogonal chemistry utility in vivo in the near future.
Collapse
Affiliation(s)
- Jianbing Wu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, 830054, P.R. China and State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, P.R. China.
| | - Tao Sun
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, 830054, P.R. China and State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, P.R. China.
| | - Chenxi Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, P.R. China.
| | - Tian Lv
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, P.R. China.
| | - Yuyang Bi
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, P.R. China.
| | - Yuan Xu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, P.R. China.
| | - Yong Ling
- School of Pharmacy, Nantong University, Nantong, 226001, P.R. China
| | - Jun Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, 830054, P.R. China and Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, P.R. China
| | - Rigang Cong
- National-certified Enterprise Technology Center, Disha Pharmaceutical Group Co., Ltd., Weihai 264205, P.R. China
| | - Yihua Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, P.R. China.
| | - Jianhua Wang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, 830054, P.R. China and Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, P.R. China
| | - Hao Wen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, 830054, P.R. China
| | - Hulin Jiang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, 830054, P.R. China and State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, P.R. China.
| | - Fei Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, P.R. China.
| | - Zhangjian Huang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, 830054, P.R. China and State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, P.R. China.
| |
Collapse
|
27
|
Pérez-López AM, Rubio-Ruiz B, Valero T, Contreras-Montoya R, Álvarez de Cienfuegos L, Sebastián V, Santamaría J, Unciti-Broceta A. Bioorthogonal Uncaging of Cytotoxic Paclitaxel through Pd Nanosheet-Hydrogel Frameworks. J Med Chem 2020; 63:9650-9659. [PMID: 32787091 PMCID: PMC7497487 DOI: 10.1021/acs.jmedchem.0c00781] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Indexed: 12/21/2022]
Abstract
The promising potential of bioorthogonal catalysis in biomedicine is inspiring incremental efforts to design strategies that regulate drug activity in living systems. To achieve this, it is not only essential to develop customized inactive prodrugs and biocompatible metal catalysts but also the right physical environment for them to interact and enable drug production under spatial and/or temporal control. Toward this goal, here, we report the first inactive precursor of the potent broad-spectrum anticancer drug paclitaxel (a.k.a. Taxol) that is stable in cell culture and labile to Pd catalysts. This new prodrug is effectively uncaged in cancer cell culture by Pd nanosheets captured within agarose and alginate hydrogels, providing a biodegradable catalytic framework to achieve controlled release of one of the most important chemotherapy drugs in medical practice. The compatibility of bioorthogonal catalysis and physical hydrogels opens up new opportunities to administer and modulate the mobility of transition metal catalysts in living environs.
Collapse
Affiliation(s)
- Ana M. Pérez-López
- Cancer
Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular
Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, U.K.
| | - Belén Rubio-Ruiz
- Cancer
Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular
Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, U.K.
| | - Teresa Valero
- Cancer
Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular
Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, U.K.
| | - Rafael Contreras-Montoya
- Departamento
de Química Orgánica, Facultad de Ciencias, Universidad de Granada, Campus de Fuentenueva s/n, Granada 18002, Spain
| | - Luis Álvarez de Cienfuegos
- Departamento
de Química Orgánica, Facultad de Ciencias, Universidad de Granada, Campus de Fuentenueva s/n, Granada 18002, Spain
| | - Víctor Sebastián
- Department
of Chemical Engineering and Environmental Technology; Instituto de
Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain
- Networking
Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-
BBN), Madrid 28029, Spain
| | - Jesús Santamaría
- Department
of Chemical Engineering and Environmental Technology; Instituto de
Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain
- Networking
Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-
BBN), Madrid 28029, Spain
| | - Asier Unciti-Broceta
- Cancer
Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular
Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, U.K.
| |
Collapse
|
28
|
Vong K, Yamamoto T, Chang TC, Tanaka K. Bioorthogonal release of anticancer drugs via gold-triggered 2-alkynylbenzamide cyclization. Chem Sci 2020; 11:10928-10933. [PMID: 34094342 PMCID: PMC8162444 DOI: 10.1039/d0sc04329j] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 08/22/2020] [Indexed: 01/10/2023] Open
Abstract
Metal-based uncaging of biomolecules has become an emerging approach for in vivo applications, which is largely due to the advantageous bioorthogonality of abiotic transition metals. Adding to the library of metal-cleavable protecting groups, this work introduces the 2-alkynylbenzamide (Ayba) moiety for the gold-triggered release of secondary amines under mild and physiological conditions. Studies were further performed to highlight some intrinsic benefits of the Ayba protecting group, which are (1) its amenable nature to derivatization for manipulating prodrug properties, and (2) its orthogonality with other commonly used transition metals like palladium and ruthenium. With a focus on highlighting its application for anticancer drug therapies, this study successfully shows that gold-triggered conversion of Ayba-protected prodrugs into bioactive anticancer drugs (i.e. doxorubicin, endoxifen) can proceed effectively in cell-based assays.
Collapse
Affiliation(s)
- Kenward Vong
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
- GlycoTargeting Research Laboratory, RIKEN Baton Zone Program 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
| | - Tomoya Yamamoto
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
| | - Tsung-Che Chang
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
| | - Katsunori Tanaka
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
- GlycoTargeting Research Laboratory, RIKEN Baton Zone Program 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
- Biofunctional Chemistry Laboratory, A. Butlerov Institute of Chemistry, Kazan Federal University 18 Kremlyovskaya Street Kazan 420008 Russia
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology 2-12-1 O-okayama Meguro-ku Tokyo 152-8552 Japan
| |
Collapse
|
29
|
Liu Y, Bai Y. Design and Engineering of Metal Catalysts for Bio-orthogonal Catalysis in Living Systems. ACS APPLIED BIO MATERIALS 2020; 3:4717-4746. [DOI: 10.1021/acsabm.0c00581] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Ying Liu
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chem/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Yugang Bai
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chem/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| |
Collapse
|
30
|
Huang R, Li CH, Cao-Milán R, He LD, Makabenta JM, Zhang X, Yu E, Rotello VM. Polymer-Based Bioorthogonal Nanocatalysts for the Treatment of Bacterial Biofilms. J Am Chem Soc 2020; 142:10723-10729. [PMID: 32464057 PMCID: PMC7339739 DOI: 10.1021/jacs.0c01758] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Bioorthogonal catalysis offers a unique strategy to modulate biological processes through the in situ generation of therapeutic agents. However, the direct application of bioorthogonal transition metal catalysts (TMCs) in complex media poses numerous challenges due to issues of limited biocompatibility, poor water solubility, and catalyst deactivation in biological environments. We report here the creation of catalytic "polyzymes", comprised of self-assembled polymer nanoparticles engineered to encapsulate lipophilic TMCs. The incorporation of catalysts into these nanoparticle scaffolds creates water-soluble constructs that provide a protective environment for the catalyst. The potential therapeutic utility of these nanozymes was demonstrated through antimicrobial studies in which a cationic nanozyme was able to penetrate into biofilms and eradicate embedded bacteria through the bioorthogonal activation of a pro-antibiotic.
Collapse
Affiliation(s)
- Rui Huang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Cheng-Hsuan Li
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Roberto Cao-Milán
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Luke D. He
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Jessa Marie Makabenta
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Xianzhi Zhang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Erlei Yu
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
- Key Laboratory for Green Process of Chemical Engineering of Xinjiang Bingtuan, School of Chemistry and Chemical Engineering, Shihezi University, Shihezi 832003, P. R. China
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| |
Collapse
|
31
|
Oliveira BL, Stenton BJ, Unnikrishnan VB, de Almeida CR, Conde J, Negrão M, Schneider FSS, Cordeiro C, Ferreira MG, Caramori GF, Domingos JB, Fior R, Bernardes GJL. Platinum-Triggered Bond-Cleavage of Pentynoyl Amide and N-Propargyl Handles for Drug-Activation. J Am Chem Soc 2020; 142:10869-10880. [PMID: 32456416 PMCID: PMC7304066 DOI: 10.1021/jacs.0c01622] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
The
ability to create ways to control drug activation at specific
tissues while sparing healthy tissues remains a major challenge. The
administration of exogenous target-specific triggers offers the potential
for traceless release of active drugs on tumor sites from antibody–drug
conjugates (ADCs) and caged prodrugs. We have developed a metal-mediated
bond-cleavage reaction that uses platinum complexes [K2PtCl4 or Cisplatin (CisPt)] for drug activation. Key to
the success of the reaction is a water-promoted activation process
that triggers the reactivity of the platinum complexes. Under these
conditions, the decaging of pentynoyl tertiary amides and N-propargyls occurs rapidly in aqueous systems. In cells,
the protected analogues of cytotoxic drugs 5-fluorouracil (5-FU) and
monomethyl auristatin E (MMAE) are partially activated by nontoxic
amounts of platinum salts. Additionally, a noninternalizing ADC built
with a pentynoyl traceless linker that features a tertiary amide protected
MMAE was also decaged in the presence of platinum salts for extracellular
drug release in cancer cells. Finally, CisPt-mediated prodrug activation
of a propargyl derivative of 5-FU was shown in a colorectal zebrafish
xenograft model that led to significant reductions in tumor size.
Overall, our results reveal a new metal-based cleavable reaction that
expands the application of platinum complexes beyond those in catalysis
and cancer therapy.
Collapse
Affiliation(s)
- Bruno L Oliveira
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Benjamin J Stenton
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - V B Unnikrishnan
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Cátia Rebelo de Almeida
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Avenida Brasilia, 1400-038 Lisboa, Portugal
| | - João Conde
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Magda Negrão
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Avenida Brasilia, 1400-038 Lisboa, Portugal
| | - Felipe S S Schneider
- Department of Chemistry, Federal University of Santa Catarina-UFSC, Campus Trindade, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Carlos Cordeiro
- Laboratório de FT-ICR e Espectrometria de Massa Estrutural, Faculdade de Ciências da Universidade de Lisboa, Campo-Grande, 1749-016 Lisboa, Portugal
| | - Miguel Godinho Ferreira
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Avenida Brasilia, 1400-038 Lisboa, Portugal.,Institute for Research on Cancer and Aging of Nice (IRCAN), Université Côte d'Azur, UMR7284 U1081 UNS, 06107 Nice, France
| | - Giovanni F Caramori
- Department of Chemistry, Federal University of Santa Catarina-UFSC, Campus Trindade, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Josiel B Domingos
- Department of Chemistry, Federal University of Santa Catarina-UFSC, Campus Trindade, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Rita Fior
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Avenida Brasilia, 1400-038 Lisboa, Portugal
| | - Gonçalo J L Bernardes
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| |
Collapse
|
32
|
Latocheski E, Dal Forno GM, Ferreira TM, Oliveira BL, Bernardes GJL, Domingos JB. Mechanistic insights into transition metal-mediated bioorthogonal uncaging reactions. Chem Soc Rev 2020; 49:7710-7729. [DOI: 10.1039/d0cs00630k] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review assesses the mechanistic aspects of transition metal-mediated uncaging reactions, with the goal of aiding the rational development of new caging groups/catalysts for chemical biology and drug-delivery applications.
Collapse
Affiliation(s)
- Eloah Latocheski
- LaCBio – Laboratory of Biomimetic Catalysis
- Department of Chemistry
- Federal University of Santa Catarina – UFSC
- 88040-900 Florianópolis
- Brazil
| | - Gean M. Dal Forno
- LaCBio – Laboratory of Biomimetic Catalysis
- Department of Chemistry
- Federal University of Santa Catarina – UFSC
- 88040-900 Florianópolis
- Brazil
| | - Thuany M. Ferreira
- LaCBio – Laboratory of Biomimetic Catalysis
- Department of Chemistry
- Federal University of Santa Catarina – UFSC
- 88040-900 Florianópolis
- Brazil
| | - Bruno L. Oliveira
- Department of Chemistry
- University of Cambridge
- CB2 1EW Cambridge
- UK
- Instituto de Medicina Molecular
| | - Gonçalo J. L. Bernardes
- Department of Chemistry
- University of Cambridge
- CB2 1EW Cambridge
- UK
- Instituto de Medicina Molecular
| | - Josiel B. Domingos
- LaCBio – Laboratory of Biomimetic Catalysis
- Department of Chemistry
- Federal University of Santa Catarina – UFSC
- 88040-900 Florianópolis
- Brazil
| |
Collapse
|
33
|
Pohorilets I, Tracey MP, LeClaire MJ, Moore EM, Lu G, Liu P, Koide K. Kinetics and Inverse Temperature Dependence of a Tsuji–Trost Reaction in Aqueous Buffer. ACS Catal 2019. [DOI: 10.1021/acscatal.9b03011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Ivanna Pohorilets
- Department of Chemistry, University of Pittsburgh 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Matthew P. Tracey
- Department of Chemistry, University of Pittsburgh 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Michael J. LeClaire
- Department of Chemistry, University of Pittsburgh 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Emily M. Moore
- Department of Chemistry, University of Pittsburgh 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Gang Lu
- Department of Chemistry, University of Pittsburgh 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Peng Liu
- Department of Chemistry, University of Pittsburgh 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Kazunori Koide
- Department of Chemistry, University of Pittsburgh 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
34
|
Sancho-Albero M, Rubio-Ruiz B, Pérez-López AM, Sebastián V, Martín-Duque P, Arruebo M, Santamaría J, Unciti-Broceta A. Cancer-derived exosomes loaded with ultrathin palladium nanosheets for targeted bioorthogonal catalysis. Nat Catal 2019; 2:864-872. [PMID: 31620674 PMCID: PMC6795537 DOI: 10.1038/s41929-019-0333-4] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 07/22/2019] [Indexed: 01/17/2023]
Abstract
The transformational impact of bioorthogonal chemistries has inspired new strategies for the in vivo synthesis of bioactive agents through non-natural means. Among these, palladium (Pd) catalysts have played a prominent role in the growing subfield of bioorthogonal catalysis by producing xenobiotics and uncaging biomolecules in living systems. However, delivering catalysts selectively to specific cell types still lags behind catalyst development. Here we have developed a bio-artificial device consisting of cancer-derived exosomes loaded with Pd catalysts by a method that enables the controlled assembly of Pd nanosheets directly inside the vesicles. This hybrid system mediates Pd-triggered dealkylation reactions in vitro and inside cells and displays preferential tropism for their progenitor cells. The use of Trojan exosomes to deliver abiotic catalysts into designated cancer cells creates the opportunity for a new targeted therapy modality: exosome-directed catalyst prodrug therapy, whose first steps are presented herein with the cell-specific release of the anticancer drug panobinostat.
Collapse
Affiliation(s)
- María Sancho-Albero
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Campus Río Ebro-Edificio I+D, c/Poeta Mariano Esquillor s/n, 50018 Zaragoza, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Belén Rubio-Ruiz
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK
| | - Ana M. Pérez-López
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK
| | - Víctor Sebastián
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Campus Río Ebro-Edificio I+D, c/Poeta Mariano Esquillor s/n, 50018 Zaragoza, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Pilar Martín-Duque
- Instituto Aragonés de Ciencias de la Salud//Fundación Araid//IIS Aragón. Centro de Investigaciones Biomédicas de Aragón, Avda San Juan Bosco 13, 50009 Zaragoza, Spain
| | - Manuel Arruebo
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Campus Río Ebro-Edificio I+D, c/Poeta Mariano Esquillor s/n, 50018 Zaragoza, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Jesús Santamaría
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Campus Río Ebro-Edificio I+D, c/Poeta Mariano Esquillor s/n, 50018 Zaragoza, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Asier Unciti-Broceta
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK
| |
Collapse
|
35
|
Wu G, Zhao T, Kang D, Zhang J, Song Y, Namasivayam V, Kongsted J, Pannecouque C, De Clercq E, Poongavanam V, Liu X, Zhan P. Overview of Recent Strategic Advances in Medicinal Chemistry. J Med Chem 2019; 62:9375-9414. [PMID: 31050421 DOI: 10.1021/acs.jmedchem.9b00359] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introducing novel strategies, concepts, and technologies that speed up drug discovery and the drug development cycle is of great importance both in the highly competitive pharmaceutical industry as well as in academia. This Perspective aims to present a "big-picture" overview of recent strategic innovations in medicinal chemistry and drug discovery.
Collapse
Affiliation(s)
- Gaochan Wu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences , Shandong University , 44 West Culture Road , 250012 Ji'nan , Shandong , P. R. China
| | - Tong Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences , Shandong University , 44 West Culture Road , 250012 Ji'nan , Shandong , P. R. China
| | - Dongwei Kang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences , Shandong University , 44 West Culture Road , 250012 Ji'nan , Shandong , P. R. China
| | - Jian Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences , Shandong University , 44 West Culture Road , 250012 Ji'nan , Shandong , P. R. China
| | - Yuning Song
- Department of Clinical Pharmacy , Qilu Hospital of Shandong University , 250012 Ji'nan , China
| | - Vigneshwaran Namasivayam
- Pharmaceutical Institute, Pharmaceutical Chemistry II , University of Bonn , 53121 Bonn , Germany
| | - Jacob Kongsted
- Department of Physics, Chemistry, and Pharmacy , University of Southern Denmark , DK-5230 Odense M , Denmark
| | - Christophe Pannecouque
- Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy , K.U. Leuven , Herestraat 49 Postbus 1043 (09.A097) , B-3000 Leuven , Belgium
| | - Erik De Clercq
- Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy , K.U. Leuven , Herestraat 49 Postbus 1043 (09.A097) , B-3000 Leuven , Belgium
| | - Vasanthanathan Poongavanam
- Department of Physics, Chemistry, and Pharmacy , University of Southern Denmark , DK-5230 Odense M , Denmark
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences , Shandong University , 44 West Culture Road , 250012 Ji'nan , Shandong , P. R. China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences , Shandong University , 44 West Culture Road , 250012 Ji'nan , Shandong , P. R. China
| |
Collapse
|
36
|
Abstract
Bioorthogonal reactions that proceed readily under physiological conditions without interference from biomolecules have found widespread application in the life sciences. Complementary to the bioorthogonal reactions that ligate two molecules, reactions that release a molecule or cleave a linker are increasingly attracting interest. Such dissociative bioorthogonal reactions have a broad spectrum of uses, for example, in controlling bio-macromolecule activity, in drug delivery, and in diagnostic assays. This review article summarizes the developed bioorthogonal reactions linked to a release step, outlines representative areas of the applications of such reactions, and discusses aspects that require further improvement.
Collapse
Affiliation(s)
- Julian Tu
- Department of Medicinal Chemistry, University of Utah, 30 S 2000 E, Salt Lake City, Utah, 84112, USA
| | - Minghao Xu
- Department of Medicinal Chemistry, University of Utah, 30 S 2000 E, Salt Lake City, Utah, 84112, USA
| | - Raphael M Franzini
- Department of Medicinal Chemistry, University of Utah, 30 S 2000 E, Salt Lake City, Utah, 84112, USA
| |
Collapse
|
37
|
Abstract
Bioorthogonal nanocatalysts in the form of 'nanozymes', are promising tools for generating imaging and therapeutic molecules in living systems. These systems use transformations developed by synthetic chemists to effect transformations that cannot be performed by cellular machinery. This emerging platform is rapidly evolving towards the creation of smart nanodevices featuring the capabilities of their enzyme prototypes, modulating catalytic activity through structure as well as chemical and physical signals. Here we describe different strategies to fabricate these nanocatalysts and their potential in diagnostic and therapeutic applications.
Collapse
|
38
|
Soldevila-Barreda JJ, Metzler-Nolte N. Intracellular Catalysis with Selected Metal Complexes and Metallic Nanoparticles: Advances toward the Development of Catalytic Metallodrugs. Chem Rev 2019; 119:829-869. [PMID: 30618246 DOI: 10.1021/acs.chemrev.8b00493] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Platinum-containing drugs (e.g., cisplatin) are among the most frequently used chemotherapeutic agents. Their tremendous success has spurred research and development of other metal-based drugs, with notable achievements. Generally, the vast majority of metal-based drug candidates in clinical and developmental stages are stoichiometric agents, i.e., each metal complex reacts only once with their biological target. Additionally, many of these metal complexes are involved in side reactions, which not only reduce the effective amount of the drug but may also cause toxicity. On a separate note, transition metal complexes and nanoparticles have a well-established history of being potent catalysts for selective molecular transformations, with examples such as the Mo- and Ru-based catalysts for metathesis reactions (Nobel Prize in 2005) or palladium catalysts for C-C bond forming reactions such as Heck, Negishi, or Suzuki reactions (Nobel Prize in 2010). Also, notably, no direct biological equivalent of these transformations exists in a biological environment such as bacteria or mammalian cells. It is, therefore, only logical that recent interest has focused on developing transition-metal based catalytic systems that are capable of performing transformations inside cells, with the aim of inducing medicinally relevant cellular changes. Because unlike in stoichiometric reactions, a catalytically active compound may turn over many substrate molecules, only very small amounts of such a catalytic metallodrug are required to achieve a desired pharmacologic effect, and therefore, toxicity and side reactions are reduced. Furthermore, performing catalytic reactions in biological systems also opens the door for new methodologies to study the behavior of biomolecules in their natural state, e.g., via in situ labeling or by increasing/depleting their concentration at will. There is, of course, an art to the choice of catalysts and reactions which have to be compatible with biological conditions, namely an aqueous, oxygen-containing environment. In this review, we aim to describe new developments that bring together the far-distant worlds of transition-metal based catalysis and metal-based drugs, in what is termed "catalytic metallodrugs". Here we will focus on transformations that have been performed on small biomolecules (such as shifting equilibria like in the NAD+/NADH or GSH/GSSG couples), on non-natural molecules such as dyes for imaging purposes, or on biomacromolecules such as proteins. Neither reactions involving release (e.g., CO) or transformation of small molecules (e.g., 1O2 production), degradation of biomolecules such as proteins, RNA or DNA nor light-induced medicinal chemistry (e.g., photodynamic therapy) are covered, even if metal complexes are centrally involved in those. In each section, we describe the (inorganic) chemistry involved, as well as selected examples of biological applications in the hope that this snapshot of a new but quickly developing field will indeed inspire novel research and unprecedented interactions across disciplinary boundaries.
Collapse
Affiliation(s)
- Joan Josep Soldevila-Barreda
- Inorganic Chemistry I-Bioinorganic Chemistry , Ruhr University Bochum , Universitätsstrasse 150 , 44780-D Bochum , Germany
| | - Nils Metzler-Nolte
- Inorganic Chemistry I-Bioinorganic Chemistry , Ruhr University Bochum , Universitätsstrasse 150 , 44780-D Bochum , Germany
| |
Collapse
|
39
|
Torres-Sánchez C, Pérez-López AM, Alqahtani MN, Unciti-Broceta A, Rubio-Ruiz B. Design and manufacture of functional catalyst-carrier structures for the bioorthogonal activation of anticancer agents. NEW J CHEM 2019. [DOI: 10.1039/c8nj05704d] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Achieving the activation of a latent prodrug via bio-orthogonal chemistry on the catalytic surface of a tailored Ti-[Pd] device.
Collapse
Affiliation(s)
- Carmen Torres-Sánchez
- Wolfson School of Mechanical
- Electrical and Manufacturing Engineering
- Loughborough University
- Loughborough
- UK
| | - Ana M. Pérez-López
- Cancer Research UK Edinburgh Centre
- Institute of Genetics and Molecular Medicine
- The University of Edinburgh
- Edinburgh EH4 2XR
- UK
| | - Mohammad N. Alqahtani
- Wolfson School of Mechanical
- Electrical and Manufacturing Engineering
- Loughborough University
- Loughborough
- UK
| | - Asier Unciti-Broceta
- Cancer Research UK Edinburgh Centre
- Institute of Genetics and Molecular Medicine
- The University of Edinburgh
- Edinburgh EH4 2XR
- UK
| | - Belén Rubio-Ruiz
- Cancer Research UK Edinburgh Centre
- Institute of Genetics and Molecular Medicine
- The University of Edinburgh
- Edinburgh EH4 2XR
- UK
| |
Collapse
|
40
|
Adam C, Pérez‐López AM, Hamilton L, Rubio‐Ruiz B, Bray TL, Sieger D, Brennan PM, Unciti‐Broceta A. Bioorthogonal Uncaging of the Active Metabolite of Irinotecan by Palladium-Functionalized Microdevices. Chemistry 2018; 24:16783-16790. [PMID: 30187973 PMCID: PMC6282958 DOI: 10.1002/chem.201803725] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Indexed: 12/20/2022]
Abstract
SN-38, the active metabolite of irinotecan, is released upon liver hydrolysis to mediate potent antitumor activity. Systemic exposure to SN-38, however, also leads to serious side effects. To reduce systemic toxicity by controlling where and when SN-38 is generated, a new prodrug was specifically designed to be metabolically stable and undergo rapid palladium-mediated activation. Blocking the phenolic OH of SN-38 with a 2,6-bis(propargyloxy)benzyl group led to significant reduction of cytotoxic activity (up to 44-fold). Anticancer properties were swiftly restored in the presence of heterogeneous palladium (Pd) catalysts to kill colorectal cancer and glioma cells, proving the efficacy of this novel masking strategy for aromatic hydroxyls. Combination with a Pd-activated 5FU prodrug augmented the antiproliferative potency of the treatment, while displaying no activity in the absence of the Pd source, which illustrates the benefit of achieving controlled release of multiple approved therapeutics-sequentially or simultaneously-by the same bioorthogonal catalyst to increase anticancer activity.
Collapse
Affiliation(s)
- Catherine Adam
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Molecular MedicineUniversity of EdinburghUK
| | - Ana M. Pérez‐López
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Molecular MedicineUniversity of EdinburghUK
| | - Lloyd Hamilton
- Centre for Neurogeneration, The Chancellor's BuildingUniversity of EdinburghUK
| | - Belén Rubio‐Ruiz
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Molecular MedicineUniversity of EdinburghUK
| | - Thomas L. Bray
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Molecular MedicineUniversity of EdinburghUK
| | - Dirk Sieger
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Molecular MedicineUniversity of EdinburghUK
- Centre for Neurogeneration, The Chancellor's BuildingUniversity of EdinburghUK
| | - Paul M. Brennan
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Molecular MedicineUniversity of EdinburghUK
- Centre for Clinical Brain SciencesUniversity of EdinburghUK
| | - Asier Unciti‐Broceta
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Molecular MedicineUniversity of EdinburghUK
| |
Collapse
|
41
|
Bray TL, Salji M, Brombin A, Pérez-López AM, Rubio-Ruiz B, Galbraith LCA, Patton EE, Leung HY, Unciti-Broceta A. Bright insights into palladium-triggered local chemotherapy. Chem Sci 2018; 9:7354-7361. [PMID: 30542538 PMCID: PMC6237126 DOI: 10.1039/c8sc02291g] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 07/16/2018] [Indexed: 12/22/2022] Open
Abstract
The incorporation of transition metal catalysts to the bioorthogonal toolbox has opened the possibility of producing supra-stoichiometric amounts of xenobiotics in living systems in a non-enzymatic fashion. For medical use, such metals could be embedded in implantable devices (i.e. heterogeneous catalyst) to "synthesize" drugs in desired locations (e.g. in a tumour) with high specificity and for extended periods of time, overcoming the useful life limitations of current local therapy modalities directed to specific organ sites (e.g. brachytherapy, controlled release systems). To translate this approach into a bona fide therapeutic option, it is essential to develop clinically-accessible implantation procedures and to understand and validate the activation process in relevant preclinical models. Herein we report the development of a novel Pd-activatable precursor of the red-fluorescent drug doxorubicin and Pd devices of optimized size and activity. Screening in state-of-the-art cancer models provided fundamental insights into the insertion protocols, safety and stability of the devices and into the prodrug distribution profile before and after activation.
Collapse
Affiliation(s)
- Thomas L Bray
- Cancer Research UK Edinburgh Centre , Institute of Genetics and Molecular Medicine , University of Edinburgh , Crewe Road South , Edinburgh EH4 2XR , UK .
| | - Mark Salji
- Institute of Cancer Sciences , University of Glasgow , Bearsden , Glasgow G61 1QH , UK .
- CRUK Beatson Institute , Bearsden , Glasgow G61 1BD , UK
| | - Alessandro Brombin
- Cancer Research UK Edinburgh Centre , Institute of Genetics and Molecular Medicine , University of Edinburgh , Crewe Road South , Edinburgh EH4 2XR , UK .
- MRC Human Genetics Unit , Institute of Genetics & Molecular Medicine , University of Edinburgh , Crewe Road South , Edinburgh EH4 2XR , UK
| | - Ana M Pérez-López
- Cancer Research UK Edinburgh Centre , Institute of Genetics and Molecular Medicine , University of Edinburgh , Crewe Road South , Edinburgh EH4 2XR , UK .
| | - Belén Rubio-Ruiz
- Cancer Research UK Edinburgh Centre , Institute of Genetics and Molecular Medicine , University of Edinburgh , Crewe Road South , Edinburgh EH4 2XR , UK .
| | - Laura C A Galbraith
- Institute of Cancer Sciences , University of Glasgow , Bearsden , Glasgow G61 1QH , UK .
- CRUK Beatson Institute , Bearsden , Glasgow G61 1BD , UK
| | - E Elizabeth Patton
- Cancer Research UK Edinburgh Centre , Institute of Genetics and Molecular Medicine , University of Edinburgh , Crewe Road South , Edinburgh EH4 2XR , UK .
- MRC Human Genetics Unit , Institute of Genetics & Molecular Medicine , University of Edinburgh , Crewe Road South , Edinburgh EH4 2XR , UK
| | - Hing Y Leung
- Institute of Cancer Sciences , University of Glasgow , Bearsden , Glasgow G61 1QH , UK .
- CRUK Beatson Institute , Bearsden , Glasgow G61 1BD , UK
| | - Asier Unciti-Broceta
- Cancer Research UK Edinburgh Centre , Institute of Genetics and Molecular Medicine , University of Edinburgh , Crewe Road South , Edinburgh EH4 2XR , UK .
| |
Collapse
|
42
|
Stenton BJ, Oliveira BL, Matos MJ, Sinatra L, Bernardes GJL. A thioether-directed palladium-cleavable linker for targeted bioorthogonal drug decaging. Chem Sci 2018; 9:4185-4189. [PMID: 29780549 PMCID: PMC5941270 DOI: 10.1039/c8sc00256h] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/02/2018] [Indexed: 01/24/2023] Open
Abstract
We describe the development of a bifunctional linker that simultaneously allows site-specific protein modification and palladium-mediated bioorthogonal decaging.
We describe the development of a bifunctional linker that simultaneously allows site-specific protein modification and palladium-mediated bioorthogonal decaging. This was enabled by a thioether binding motif in the propargyl carbamate linker and a readily available palladium complex. We demonstrate the efficiency of this reaction by controlled drug release from a PEGylated doxorubicin prodrug in cancer cells. The linker can be easily installed into cysteine bearing proteins which we demonstrated for the construction of an anti-HER2 nanobody–drug conjugate. Targeted delivery of the nanobody drug conjugate showed effective cell killing in HER2+ cells upon palladium-mediated decaging.
Collapse
Affiliation(s)
- Benjamin J Stenton
- Department of Chemistry , University of Cambridge , Lensfield Road , CB2 1EW Cambridge , UK .
| | - Bruno L Oliveira
- Department of Chemistry , University of Cambridge , Lensfield Road , CB2 1EW Cambridge , UK .
| | - Maria J Matos
- Department of Chemistry , University of Cambridge , Lensfield Road , CB2 1EW Cambridge , UK .
| | - Laura Sinatra
- Department of Chemistry , University of Cambridge , Lensfield Road , CB2 1EW Cambridge , UK .
| | - Gonçalo J L Bernardes
- Department of Chemistry , University of Cambridge , Lensfield Road , CB2 1EW Cambridge , UK . .,Instituto de Medicina Molecular , Faculdade de Medicina da Universidade de Lisboa , Av. Prof. Egas Moniz , 1649-028 Lisboa , Portugal .
| |
Collapse
|
43
|
Lv T, Wu J, Kang F, Wang T, Wan B, Lu JJ, Zhang Y, Huang Z. Synthesis and Evaluation of O2-Derived Diazeniumdiolates Activatable via Bioorthogonal Chemistry Reactions in Living Cells. Org Lett 2018; 20:2164-2167. [DOI: 10.1021/acs.orglett.8b00423] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Tian Lv
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Jianbing Wu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Fenghua Kang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Tingting Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Boheng Wan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yihua Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Zhangjian Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, P. R. China
| |
Collapse
|
44
|
Liu Y, Pujals S, Stals PJM, Paulöhrl T, Presolski SI, Meijer EW, Albertazzi L, Palmans ARA. Catalytically Active Single-Chain Polymeric Nanoparticles: Exploring Their Functions in Complex Biological Media. J Am Chem Soc 2018; 140:3423-3433. [PMID: 29457449 PMCID: PMC5997400 DOI: 10.1021/jacs.8b00122] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Indexed: 01/01/2023]
Abstract
Dynamic single-chain polymeric nanoparticles (SCPNs) are intriguing, bioinspired architectures that result from the collapse or folding of an individual polymer chain into a nanometer-sized particle. Here we present a detailed biophysical study on the behavior of dynamic SCPNs in living cells and an evaluation of their catalytic functionality in such a complex medium. We first developed a number of delivery strategies that allowed the selective localization of SCPNs in different cellular compartments. Live/dead tests showed that the SCPNs were not toxic to cells while spectral imaging revealed that SCPNs provide a structural shielding and reduced the influence from the outer biological media. The ability of SCPNs to act as catalysts in biological media was first assessed by investigating their potential for reactive oxygen species generation. With porphyrins covalently attached to the SCPNs, singlet oxygen was generated upon irradiation with light, inducing spatially controlled cell death. In addition, Cu(I)- and Pd(II)-based SCPNs were prepared and these catalysts were screened in vitro and studied in cellular environments for the carbamate cleavage reaction of rhodamine-based substrates. This is a model reaction for the uncaging of bioactive compounds such as cytotoxic drugs for catalysis-based cancer therapy. We observed that the rate of the deprotection depends on both the organometallic catalysts and the nature of the protective group. The rate reduces from in vitro to the biological environment, indicating a strong influence of biomolecules on catalyst performance. The Cu(I)-based SCPNs in combination with the dimethylpropargyloxycarbonyl protective group showed the best performances both in vitro and in biological environment, making this group promising in biomedical applications.
Collapse
Affiliation(s)
- Yiliu Liu
- Laboratory
for Macromolecular and Organic Chemistry and Institute for Complex
Molecular Systems, Eindhoven University
of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Sílvia Pujals
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Carrer de Baldiri Reixac 15-21, 08028 Barcelona, Spain
| | - Patrick J. M. Stals
- Laboratory
for Macromolecular and Organic Chemistry and Institute for Complex
Molecular Systems, Eindhoven University
of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Thomas Paulöhrl
- Laboratory
for Macromolecular and Organic Chemistry and Institute for Complex
Molecular Systems, Eindhoven University
of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Stanislav I. Presolski
- Laboratory
for Macromolecular and Organic Chemistry and Institute for Complex
Molecular Systems, Eindhoven University
of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - E. W. Meijer
- Laboratory
for Macromolecular and Organic Chemistry and Institute for Complex
Molecular Systems, Eindhoven University
of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Lorenzo Albertazzi
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Carrer de Baldiri Reixac 15-21, 08028 Barcelona, Spain
| | - Anja R. A. Palmans
- Laboratory
for Macromolecular and Organic Chemistry and Institute for Complex
Molecular Systems, Eindhoven University
of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
45
|
Miguel-Ávila J, Tomás-Gamasa M, Olmos A, Pérez PJ, Mascareñas JL. Discrete Cu(i) complexes for azide-alkyne annulations of small molecules inside mammalian cells. Chem Sci 2018; 9:1947-1952. [PMID: 29675241 PMCID: PMC5892125 DOI: 10.1039/c7sc04643j] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/15/2018] [Indexed: 12/31/2022] Open
Abstract
The archetype reaction of "click" chemistry, namely, the copper-promoted azide-alkyne cycloaddition (CuAAC), has found an impressive number of applications in biological chemistry. However, methods for promoting intermolecular annulations of exogenous, small azides and alkynes in the complex interior of mammalian cells, are essentially unknown. Herein we demonstrate that isolated, well-defined copper(i)-tris(triazolyl) complexes featuring designed ligands can readily enter mammalian cells and promote intracellular CuAAC annulations of small, freely diffusible molecules. In addition to simplifying protocols and avoiding the addition of "non-innocent" reductants, the use of these premade copper complexes leads to more efficient processes than with the alternative, in situ made copper species prepared from Cu(ii) sources, tris(triazole) ligands and sodium ascorbate. Under the reaction conditions, the well-defined copper complexes exhibit very good cell penetration properties, and do not present significant toxicities.
Collapse
Affiliation(s)
- Joan Miguel-Ávila
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) , Departamento de Química Orgánica , Universidade de Santiago de Compostela , 15782 Santiago de Compostela , Spain .
| | - María Tomás-Gamasa
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) , Departamento de Química Orgánica , Universidade de Santiago de Compostela , 15782 Santiago de Compostela , Spain .
| | - Andrea Olmos
- Laboratorio de Catálisis Homogénea , Unidad Asociada al CSIC , CIQSO-Centro de Investigación en Química Sostenible , Departamento de Química , Universidad de Huelva , Campus de El Carmen s/n , 21007 Huelva , Spain .
| | - Pedro J Pérez
- Laboratorio de Catálisis Homogénea , Unidad Asociada al CSIC , CIQSO-Centro de Investigación en Química Sostenible , Departamento de Química , Universidad de Huelva , Campus de El Carmen s/n , 21007 Huelva , Spain .
| | - José L Mascareñas
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) , Departamento de Química Orgánica , Universidade de Santiago de Compostela , 15782 Santiago de Compostela , Spain .
| |
Collapse
|
46
|
Pekarik V, Peskova M, Guran R, Novacek J, Heger Z, Tripsianes K, Kumar J, Adam V. Visualization of stable ferritin complexes with palladium, rhodium and iridium nanoparticles detected by their catalytic activity in native polyacrylamide gels. Dalton Trans 2018; 46:13690-13694. [PMID: 28971191 DOI: 10.1039/c7dt02818k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The reductive discoloration of azo dye, Congo red, catalyzed by noble metal nanoparticles was used to visualize protein-metal complexes in native polyacrylamide gels after counterstaining with Coomassie blue. This technique was used to characterize the synthesis of palladium, rhodium and iridium nanoparticles encapsulated in Pyrococcus furiosus ferritin.
Collapse
Affiliation(s)
- Vladimir Pekarik
- Institute of Physiology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Rubio-Ruiz B, Pérez-López AM, Bray TL, Lee M, Serrels A, Prieto M, Arruebo M, Carragher NO, Sebastián V, Unciti-Broceta A. High-Precision Photothermal Ablation Using Biocompatible Palladium Nanoparticles and Laser Scanning Microscopy. ACS APPLIED MATERIALS & INTERFACES 2018; 10:3341-3348. [PMID: 29320154 PMCID: PMC5799879 DOI: 10.1021/acsami.7b17282] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Herein, we report a straightforward method for the scalable preparation of Pd nanoparticles (Pd-NPs) with reduced inherent cytotoxicity and high photothermal conversion capacity. These Pd-NPs are rapidly taken up by cells and able to kill labeled cancer cells upon short exposure to near-infrared (NIR) light. Following cell treatment with Pd-NPs, ablated areas were patterned with high precision by laser scanning microscopy, allowing one to perform cell migration assays with unprecedented accuracy. Using coherent Raman microscopy, cells containing Pd-NPs were simultaneously ablated and imaged. This novel methodology was combined with intravital imaging to mediate microablation of cancerous tissue in tumor xenografts in mice.
Collapse
Affiliation(s)
- Belén Rubio-Ruiz
- Cancer Research
UK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | - Ana M. Pérez-López
- Cancer Research
UK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | - Thomas L. Bray
- Cancer Research
UK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | - Martin Lee
- Cancer Research
UK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | - Alan Serrels
- MRC Centre for Inflammation Research, Queen’s Medical Research
Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Martín Prieto
- Department of Chemical Engineering, Aragon
Institute of Nanoscience (INA), University
of Zaragoza, Campus Río
Ebro-Edificio I+D, c/Poeta Mariano Esquillor s/n, 50018 Zaragoza, Spain
| | - Manuel Arruebo
- Department of Chemical Engineering, Aragon
Institute of Nanoscience (INA), University
of Zaragoza, Campus Río
Ebro-Edificio I+D, c/Poeta Mariano Esquillor s/n, 50018 Zaragoza, Spain
- Networking
Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Neil O. Carragher
- Cancer Research
UK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | - Víctor Sebastián
- Department of Chemical Engineering, Aragon
Institute of Nanoscience (INA), University
of Zaragoza, Campus Río
Ebro-Edificio I+D, c/Poeta Mariano Esquillor s/n, 50018 Zaragoza, Spain
- Networking
Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Asier Unciti-Broceta
- Cancer Research
UK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
- E-mail: . Phone: 0044 131 6518500
| |
Collapse
|
48
|
Pérez‐López AM, Rubio‐Ruiz B, Sebastián V, Hamilton L, Adam C, Bray TL, Irusta S, Brennan PM, Lloyd‐Jones GC, Sieger D, Santamaría J, Unciti‐Broceta A. Gold-Triggered Uncaging Chemistry in Living Systems. Angew Chem Int Ed Engl 2017; 56:12548-12552. [PMID: 28699691 PMCID: PMC5655737 DOI: 10.1002/anie.201705609] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/10/2017] [Indexed: 02/02/2023]
Abstract
Recent advances in bioorthogonal catalysis are increasing the capacity of researchers to manipulate the fate of molecules in complex biological systems. A bioorthogonal uncaging strategy is presented, which is triggered by heterogeneous gold catalysis and facilitates the activation of a structurally diverse range of therapeutics in cancer cell culture. Furthermore, this solid-supported catalytic system enabled locally controlled release of a fluorescent dye into the brain of a zebrafish for the first time, offering a novel way to modulate the activity of bioorthogonal reagents in the most fragile and complex organs.
Collapse
Affiliation(s)
- Ana M. Pérez‐López
- Cancer Research (UK) Edinburgh CentreMRC Institute of Genetics and Molecular MedicineUniversity of EdinburghUK
| | - Belén Rubio‐Ruiz
- Cancer Research (UK) Edinburgh CentreMRC Institute of Genetics and Molecular MedicineUniversity of EdinburghUK
| | - Víctor Sebastián
- Department of Chemical Engineering and Environmental Technology and Institute of Nanosciences of Aragon (INA)University of ZaragozaSpain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN28029MadridSpain
| | - Lloyd Hamilton
- Centre for Neurogeneration, The Chancellor's BuildingUniversity of EdinburghUK
| | - Catherine Adam
- Cancer Research (UK) Edinburgh CentreMRC Institute of Genetics and Molecular MedicineUniversity of EdinburghUK
| | - Thomas L. Bray
- Cancer Research (UK) Edinburgh CentreMRC Institute of Genetics and Molecular MedicineUniversity of EdinburghUK
| | - Silvia Irusta
- Department of Chemical Engineering and Environmental Technology and Institute of Nanosciences of Aragon (INA)University of ZaragozaSpain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN28029MadridSpain
| | - Paul M. Brennan
- Cancer Research (UK) Edinburgh CentreMRC Institute of Genetics and Molecular MedicineUniversity of EdinburghUK
- Centre for Clinical Brain SciencesUniversity of EdinburghUK
| | - Guy C. Lloyd‐Jones
- School of Chemistry, King's Buildings, West Mains RoadUniversity of EdinburghUK
| | - Dirk Sieger
- Centre for Neurogeneration, The Chancellor's BuildingUniversity of EdinburghUK
| | - Jesús Santamaría
- Department of Chemical Engineering and Environmental Technology and Institute of Nanosciences of Aragon (INA)University of ZaragozaSpain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN28029MadridSpain
| | - Asier Unciti‐Broceta
- Cancer Research (UK) Edinburgh CentreMRC Institute of Genetics and Molecular MedicineUniversity of EdinburghUK
| |
Collapse
|
49
|
Du B, Li D, Wang J, Wang E. Designing metal-contained enzyme mimics for prodrug activation. Adv Drug Deliv Rev 2017; 118:78-93. [PMID: 28412325 DOI: 10.1016/j.addr.2017.04.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 03/22/2017] [Accepted: 04/04/2017] [Indexed: 01/09/2023]
Abstract
Enzyme-activated prodrug therapy (EAPT) is a widely-used and effective treatment method for cancer by converting prodrugs into drugs at the demanded time and space, whose key step is prodrug activation. Traditional prodrug activations are mostly dependent on natural enzymes, which are unstable, expensive and hard to be functionalized. The emerging enzyme mimics, especially the metal-contained enzyme mimics (MEMs), provide a potential chance for improving the traditional EAPT because of their high stability, low cost and easiness of preparation and functionalization. The existing MEMs can be classified into three categories: catalytic core-scaffold MEM (csMEM), nanoparticle MEM (npMEMs) and metal-organic framework (MOF) MEM (mofMEM). These MEMs can mimic diverse functions corresponding to natural enzymes, and some of which are potentially used in prodrug activation, such as DNase, RNase, carbonate esterase, etc. In this review, we briefly summarize the MEMs according to their structure and composition, and highlight the successful and potential applications for prodrug activation mediated by hydrolase-like and oxidoreductase-like MEMs.
Collapse
|
50
|
Pérez‐López AM, Rubio‐Ruiz B, Sebastián V, Hamilton L, Adam C, Bray TL, Irusta S, Brennan PM, Lloyd‐Jones GC, Sieger D, Santamaría J, Unciti‐Broceta A. Gold‐Triggered Uncaging Chemistry in Living Systems. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201705609] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Ana M. Pérez‐López
- Cancer Research (UK) Edinburgh Centre MRC Institute of Genetics and Molecular Medicine University of Edinburgh UK
| | - Belén Rubio‐Ruiz
- Cancer Research (UK) Edinburgh Centre MRC Institute of Genetics and Molecular Medicine University of Edinburgh UK
| | - Víctor Sebastián
- Department of Chemical Engineering and Environmental Technology and Institute of Nanosciences of Aragon (INA) University of Zaragoza Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN 28029 Madrid Spain
| | - Lloyd Hamilton
- Centre for Neurogeneration, The Chancellor's Building University of Edinburgh UK
| | - Catherine Adam
- Cancer Research (UK) Edinburgh Centre MRC Institute of Genetics and Molecular Medicine University of Edinburgh UK
| | - Thomas L. Bray
- Cancer Research (UK) Edinburgh Centre MRC Institute of Genetics and Molecular Medicine University of Edinburgh UK
| | - Silvia Irusta
- Department of Chemical Engineering and Environmental Technology and Institute of Nanosciences of Aragon (INA) University of Zaragoza Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN 28029 Madrid Spain
| | - Paul M. Brennan
- Cancer Research (UK) Edinburgh Centre MRC Institute of Genetics and Molecular Medicine University of Edinburgh UK
- Centre for Clinical Brain Sciences University of Edinburgh UK
| | - Guy C. Lloyd‐Jones
- School of Chemistry, King's Buildings, West Mains Road University of Edinburgh UK
| | - Dirk Sieger
- Centre for Neurogeneration, The Chancellor's Building University of Edinburgh UK
| | - Jesús Santamaría
- Department of Chemical Engineering and Environmental Technology and Institute of Nanosciences of Aragon (INA) University of Zaragoza Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN 28029 Madrid Spain
| | - Asier Unciti‐Broceta
- Cancer Research (UK) Edinburgh Centre MRC Institute of Genetics and Molecular Medicine University of Edinburgh UK
| |
Collapse
|