1
|
Li J, Tang Y, Lu G, Liu Q, Guo Y, Wang J, Xiao M, Gao T, Zhang X, Gu J. Elevation of p53 sensitizes obese kidney to adriamycin-induced aberrant lipid homeostasis via repressing HNF4α-mediated FGF21 sensitivity. J Adv Res 2025; 72:165-180. [PMID: 39019110 DOI: 10.1016/j.jare.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/22/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024] Open
Abstract
INTRODUCTION Lipid metabolism disorders have been confirmed to be closely related to kidney injury caused by adriamycin (ADR) and obesity, respectively. However, it has not been explored whether lipid metabolism disorders appear progressively more severe after ADR-based chemotherapy in the obese state, and the specific molecular mechanism needs to be further clarified. OBJECTIVES This study was designed to examine the role of p53-fibroblast growth factor 21 (FGF21) axis in ADR-induced renal injury aggravated by high-fat diet (HFD). METHODS We engineered Fgf21 KO mice and used long-term (4 months) and short-term (0.5 months) HFD feeding, and ADR-injected mice, as well as STZ-induced type 1 diabetic mice and type 2 (db/db) diabetic mice to produce an in vivo model of nephrotoxicity. The specific effects of p53/FGF21 on the regulation of lipid metabolism disorders and its downstream mediators in kidney were subsequently elucidated using a combination of functional and pathological analysis, RNA-sequencing, molecular biology, and in vitro approaches. RESULTS Long-term HFD feeding mice exhibited compromised effects of FGF21 on alleviation of renal dysfunction and lipid accumulation following ADR administration. However, these impairments were reversed by p53 inhibitor (pifithrin-α, PFT-α). PFT-α sensitized FGF21 actions in kidney tissues, while knockout of Fgf21 impaired the protective effects of PFT-α on lipid metabolism. Mechanistically, p53 impaired the renal expression of FGF receptor-1 (FGFR1) and thereby developed gradually into FGF21 resistance via inhibiting hepatocyte nuclear factor 4 alpha (HNF4α)-mediated transcriptional activation of Fgfr1. More importantly, exogenous supplementation of FGF21 or PFT-α could not only alleviate ADR-induced lipid metabolism disorder aggravated by HFD, but also reduce lipid accumulation caused by diabetic nephropathy. CONCLUSION Given the difficulties in developing the long-acting recombinant FGF21 analogs for therapeutic applications, sensitizing obesity-impaired FGF21 actions by suppression of p53 might be a therapeutic strategy for maintaining renal metabolic homeostasis during chemotherapy.
Collapse
Affiliation(s)
- Jiahao Li
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yufeng Tang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Guangping Lu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Qingbo Liu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yuanfang Guo
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Mengjie Xiao
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Ting Gao
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiaohui Zhang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Junlian Gu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
2
|
Zhou Z, Sun Y, Pang J, Long YQ. Advances in the Delivery, Activation and Therapeutics Applications of Bioorthogonal Prodrugs. Med Res Rev 2025; 45:887-908. [PMID: 39692238 DOI: 10.1002/med.22095] [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: 03/08/2023] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/19/2024]
Abstract
Traditional prodrug strategies have been leveraged to overcome many inherent drawbacks of active native drugs in the drug research and development. However, endogenous stimuli such as specific microenvironment or enzymes are relied on to achieve the prodrug activation, resulting in unintended drug release and systemic toxicity. Alternatively, bioorthogonal cleavage reaction-enabled bioorthogonal prodrugs activation via exogenous triggers has emerged as a valuable approach, featuring spatiotemporally controlled drug release. Such bioorthogonal prodrug strategies would ensure targeted drug delivery and/or in situ generation, further circumventing systemic toxicity or premature elimination of active drugs. In recent years, metal-free bioorthogonal cleavage reactions with fast kinetics have boomed in the bioorthogonal prodrug design. Meanwhile, transition-metal-catalyzed and photocatalytic deprotection reactions have also been developed to trigger prodrug activation in biological systems. Besides traditional small molecule prodrugs, gasotransmitters have been successfully delivered to specific organelles or cells via bioorthogonal reactions, and nanosystems have been devised into bioorthogonal triggers as well. Herein, we present an overview of the latest advances in these bioorthogonally-uncaged prodrugs, focused on the delivery, activation and therapeutics applications.
Collapse
Affiliation(s)
- Zhou Zhou
- Department of Medicinal Chemistry, Laboratory of Medicinal Chemical Biology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yuanjun Sun
- Department of Medicinal Chemistry, Laboratory of Medicinal Chemical Biology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, China
| | - Jing Pang
- Department of Medicinal Chemistry, Laboratory of Medicinal Chemical Biology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, China
| | - Ya-Qiu Long
- Department of Medicinal Chemistry, Laboratory of Medicinal Chemical Biology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, China
| |
Collapse
|
3
|
Garcia-Peiro JI, Ortega-Liebana MC, Adam C, Lorente-Macías Á, Travnickova J, Patton EE, Guerrero-López P, Garcia-Aznar JM, Hueso JL, Santamaria J, Unciti-Broceta A. Dendritic Platinum Nanoparticles Shielded by Pt-S PEGylation as Intracellular Reactors for Bioorthogonal Uncaging Chemistry. Angew Chem Int Ed Engl 2025; 64:e202424037. [PMID: 39813113 DOI: 10.1002/anie.202424037] [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: 12/09/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 01/18/2025]
Abstract
Beyond their classical role as cytotoxics, Platinum (Pt) coordination complexes recently joined the selected group of transition metals capable of performing bioorthogonal reactions in living environments. To minimize their reactivity towards nucleophiles, which limit their catalytic performance, we investigated the use of Pt(0) with different forms, sizes and surface functionalization. We report herein the development of PEGylated Pt nanodendrites with the capacity to activate prodyes and prodrugs in cell culture and in vivo. Their dendritic morphology together with their surface shielding through Pt-S-bonded PEGylation synergistically contributed to create catalytic nanoreactors compatible with the highly-crowded and reductive environment of the cell cytoplasm, thereby facilitating in situ bioorthogonal drug uncaging in cancer cells in 2D and 3D culture, including in microfluidic systems, and xenografted in zebrafish.
Collapse
Affiliation(s)
- Jose I Garcia-Peiro
- Instituto de Nanociencia y Materiales de Aragon (INMA), CSIC-Universidad de Zaragoza, Edificio I+D, C/Poeta Mariano Esquillor s/n, 50018, Zaragoza, Spain
- Department of Chemical and Environmental Engineering, University of Zaragoza, Campus Rio Ebro, C/María de Luna, 3, 50018, Zaragoza, Spain
- Networking Research Center in Biomaterials, Bioengineering and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Instituto de Investigación Sanitaria (IIS) de Aragón, Avenida San Juan Bosco, 13, 50009, Zaragoza, Spain
| | - M Carmen Ortega-Liebana
- Edinburgh Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
- Department of Medicinal and Organic Chemistry and Unit of Excellence in Chemistry Applied to Biomedicine and Environment, Faculty of Pharmacy, Campus Cartuja s/n, University of Granada, 18071, Granada, Spain
- GENYO, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avda. Ilustración 114, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Catherine Adam
- Edinburgh Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Álvaro Lorente-Macías
- Edinburgh Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Jana Travnickova
- Edinburgh Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - E Elizabeth Patton
- Edinburgh Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Paula Guerrero-López
- Instituto de Investigación Sanitaria (IIS) de Aragón, Avenida San Juan Bosco, 13, 50009, Zaragoza, Spain
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3 A), University of Zaragoza, Mariano Esquillor s/n, 50018, Zaragoza, Spain
| | - J Manuel Garcia-Aznar
- Instituto de Investigación Sanitaria (IIS) de Aragón, Avenida San Juan Bosco, 13, 50009, Zaragoza, Spain
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3 A), University of Zaragoza, Mariano Esquillor s/n, 50018, Zaragoza, Spain
| | - Jose L Hueso
- Instituto de Nanociencia y Materiales de Aragon (INMA), CSIC-Universidad de Zaragoza, Edificio I+D, C/Poeta Mariano Esquillor s/n, 50018, Zaragoza, Spain
- Department of Chemical and Environmental Engineering, University of Zaragoza, Campus Rio Ebro, C/María de Luna, 3, 50018, Zaragoza, Spain
- Networking Research Center in Biomaterials, Bioengineering and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Instituto de Investigación Sanitaria (IIS) de Aragón, Avenida San Juan Bosco, 13, 50009, Zaragoza, Spain
- Escuela Politécnica Superior, Universidad de Zaragoza. Crta. de Cuarte s/n, 22071, Huesca, Spain
| | - Jesus Santamaria
- Instituto de Nanociencia y Materiales de Aragon (INMA), CSIC-Universidad de Zaragoza, Edificio I+D, C/Poeta Mariano Esquillor s/n, 50018, Zaragoza, Spain
- Department of Chemical and Environmental Engineering, University of Zaragoza, Campus Rio Ebro, C/María de Luna, 3, 50018, Zaragoza, Spain
- Networking Research Center in Biomaterials, Bioengineering and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Instituto de Investigación Sanitaria (IIS) de Aragón, Avenida San Juan Bosco, 13, 50009, Zaragoza, Spain
| | - Asier Unciti-Broceta
- Edinburgh Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| |
Collapse
|
4
|
Lin Y, Hashimoto R, Chang TC, Tanaka K. Synthesis of phenanthridine derivatives by a water-compatible gold-catalyzed hydroamination. Bioorg Med Chem 2024; 113:117928. [PMID: 39299083 DOI: 10.1016/j.bmc.2024.117928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
Since transition-metal-catalyzed reactions are one of the most powerful and direct approaches for the synthesis of organic molecules, translating them to biological systems for biomedical applications is an emerging field. The manipulation of transition metal reactions in biological settings for uncaging prodrugs and synthesizing bioactive drugs has been widely studied. To expand the toolbox of transition-metal-mediated prodrug strategy, this work introduces the 2'-alkynl-biphenylamine precursors for the synthesis of phenanthridine derivatives using a water-compatible gold-catalyzed hydroamination under mild conditions. Moreover, the structure-reactivity relationship revealed that the nucleophilicity of the amine group in the precursor was critical for facilitating the gold-catalyzed synthesis of phenanthridine derivatives. The research shows the potential to be used for phenanthridine-based prodrug designs in an aqueous solution.
Collapse
Affiliation(s)
- Yixuan Lin
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
| | - Riichi Hashimoto
- 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; Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro-ku, Tokyo 152-8552, Japan.
| |
Collapse
|
5
|
van de L'Isle M, Croke S, Valero T, Unciti‐Broceta A. Development of Biocompatible Cu(I)-Microdevices for Bioorthogonal Uncaging and Click Reactions. Chemistry 2024; 30:e202400611. [PMID: 38512657 PMCID: PMC11497292 DOI: 10.1002/chem.202400611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/21/2024] [Accepted: 03/21/2024] [Indexed: 03/23/2024]
Abstract
Transition-metal-catalyzed bioorthogonal reactions emerged a decade ago as a novel strategy to implement spatiotemporal control over enzymatic functions and pharmacological interventions. The use of this methodology in experimental therapy is driven by the ambition of improving the tolerability and PK properties of clinically-used therapeutic agents. The preclinical potential of bioorthogonal catalysis has been validated in vitro and in vivo with the in situ generation of a broad range of drugs, including cytotoxic agents, anti-inflammatory drugs and anxiolytics. In this article, we report our investigations towards the preparation of solid-supported Cu(I)-microdevices and their application in bioorthogonal uncaging and click reactions. A range of ligand-functionalized polymeric devices and off-on Cu(I)-sensitive sensors were developed and tested under conditions compatible with life. Last, we present a preliminary exploration of their use for the synthesis of PROTACs through CuAAC assembly of two heterofunctional mating units.
Collapse
Affiliation(s)
- Melissa van de L'Isle
- Edinburgh Cancer ResearchInstitute of Genetics & CancerUniversity of EdinburghCrewe Road SouthEdinburghEH4 2XRUK
| | - Stephen Croke
- Edinburgh Cancer ResearchInstitute of Genetics & CancerUniversity of EdinburghCrewe Road SouthEdinburghEH4 2XRUK
| | - Teresa Valero
- Edinburgh Cancer ResearchInstitute of Genetics & CancerUniversity of EdinburghCrewe Road SouthEdinburghEH4 2XRUK
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of Chemistry applied to Biomedicine and the EnvironmentFaculty of PharmacyUniversity of GranadaCampus de Cartuja s/n18071GranadaSpain
- GENYOCentre for Genomics and Oncological ResearchPfizer/University of Granada/Andalusian Regional GovernmentAvda. Ilustración 11418016GranadaSpain
- Instituto de Investigación Biosanitaria ibs.GRANADAGranadaSpain
| | - Asier Unciti‐Broceta
- Edinburgh Cancer ResearchInstitute of Genetics & CancerUniversity of EdinburghCrewe Road SouthEdinburghEH4 2XRUK
| |
Collapse
|
6
|
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
|
7
|
Liu L, Zhang X, Fedeli S, Cicek YA, Ndugire W, Rotello VM. Controlled Bio-Orthogonal Catalysis Using Nanozyme-Protein Complexes via Modulation of Electrostatic Interactions. MATERIALS (BASEL, SWITZERLAND) 2024; 17:1507. [PMID: 38612022 PMCID: PMC11012815 DOI: 10.3390/ma17071507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024]
Abstract
Bio-orthogonal chemistry provides a powerful tool for drug delivery systems due to its ability to generate therapeutic agents in situ, minimizing off-target effects. Bio-orthogonal transition metal catalysts (TMCs) with stimuli-responsive properties offer possibilities for controllable catalysis due to their spatial-, temporal-, and dosage-controllable properties. In this paper, we fabricated a stimuli-responsive bio-orthogonal catalysis system based on an enhanced green fluorescent protein (EGFP)-nanozyme (NZ) complex (EGFP-NZ). Regulation of the catalytic properties of the EGFP-NZ complex was directly achieved by modulating the ionic strength of the solution. The dielectric screening introduced by salt ions allows the dissociation of the EGFP-NZ complex, increasing the access of substrate to the active site of the NZs and concomitantly increasing nanozyme activity. The change in catalytic rate of the NZ/EGFP = 1:1 complex was positively correlated with salt concentration from 0 mM to 150 mM.
Collapse
Affiliation(s)
| | | | | | | | | | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA; (L.L.); (X.Z.); (S.F.); (Y.A.C.)
| |
Collapse
|
8
|
Ma Y, Zhou Y, Long J, Sun Q, Luo Z, Wang W, Hou T, Yin L, Zhao L, Peng J, Ding Y. A High-Efficiency Bioorthogonal Tumor-Membrane Reactor for In Situ Selective and Sustained Prodrug Activation. Angew Chem Int Ed Engl 2024; 63:e202318372. [PMID: 38205971 DOI: 10.1002/anie.202318372] [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: 11/30/2023] [Revised: 12/28/2023] [Accepted: 01/09/2024] [Indexed: 01/12/2024]
Abstract
The site-specific activation of bioorthogonal prodrugs has provided great opportunities for reducing the severe side effects of chemotherapy. However, the precise control of activation location, sustained drug production at the target site, and high bioorthogonal reaction efficiency in vivo remain great challenges. Here, we propose the construction of tumor cell membrane reactors in vivo to solve the above problems. Specifically, tumor-targeted liposomes with efficient membrane fusion capabilities are generated to install the bioorthogonal trigger, the amphiphilic tetrazine derivative, on the surface of tumor cells. These predecorated tumor cells act as many living reactors, transforming the tumor into a "drug factory" that in situ activates an externally delivered bioorthogonal prodrug, for example intratumorally injected transcyclooctene-caged doxorubicin. In contrast to the rapid elimination of cargo that is encapsulated and delivered by liposomes, these reactors permit stable retention of bioorthogonal triggers in tumor for 96 h after a single dose of liposomes via intravenous injection, allowing sustained generation of doxorubicin. Interestingly, an additional supplement of liposomes will compensate for the trigger consumed by the reaction and significantly improve the efficiency of the local reaction. This strategy provides a solution to the efficacy versus safety dilemma of tumor chemotherapy.
Collapse
Affiliation(s)
- Yu Ma
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 211112, China
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yunyun Zhou
- State Key Laboratory of Natural Medicine, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Jiaqin Long
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 211112, China
| | - Qi Sun
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 211112, China
| | - Zijiang Luo
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 211112, China
| | - Wenjie Wang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 211112, China
| | - Ting Hou
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 211112, China
| | - Li Yin
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 211112, China
| | - Lingzhi Zhao
- State Key Laboratory of Natural Medicine, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Juanjuan Peng
- State Key Laboratory of Natural Medicine, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Ya Ding
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 211112, China
| |
Collapse
|
9
|
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
|
10
|
Deng L, Sathyan A, Adam C, Unciti-Broceta A, Sebastian V, Palmans ARA. Enhanced Efficiency of Pd(0)-Based Single Chain Polymeric Nanoparticles for in Vitro Prodrug Activation by Modulating the Polymer's Microstructure. NANO LETTERS 2024; 24:2242-2249. [PMID: 38346395 PMCID: PMC10885199 DOI: 10.1021/acs.nanolett.3c04466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Bioorthogonal catalysis employing transition metal catalysts is a promising strategy for the in situ synthesis of imaging and therapeutic agents in biological environments. The transition metal Pd has been widely used as a bioorthogonal catalyst, but bare Pd poses challenges in water solubility and catalyst stability in cellular environments. In this work, Pd(0) loaded amphiphilic polymeric nanoparticles are applied to shield Pd in the presence of living cells for the in situ generation of a fluorescent dye and anticancer drugs. Pd(0) loaded polymeric nanoparticles prepared by the reduction of the corresponding Pd(II)-polymeric nanoparticles are highly active in the deprotection of pro-rhodamine dye and anticancer prodrugs, giving significant fluorescence enhancement and toxigenic effects, respectively, in HepG2 cells. In addition, we show that the microstructure of the polymeric nanoparticles for scaffolding Pd plays a critical role in tuning the catalytic efficiency, with the use of the ligand triphenylphosphine as a key factor for improving the catalyst stability in biological environments.
Collapse
Affiliation(s)
- Linlin Deng
- Laboratory for Macromolecular and Organic Chemistry, Department of Chemical Engineering and Chemistry, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Anjana Sathyan
- Laboratory for Macromolecular and Organic Chemistry, Department of Chemical Engineering and Chemistry, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Catherine Adam
- Edinburgh Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | - Asier Unciti-Broceta
- Edinburgh Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | - Víctor Sebastian
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain
- Department of Chemical and Environmental Engineering, Universidad de Zaragoza, Campus Rio Ebro, 50018 Zaragoza, Spain
- Laboratorio de Microscopías Avanzadas, Universidad de Zaragoza, 50018 Zaragoza, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Anja R A Palmans
- Laboratory for Macromolecular and Organic Chemistry, Department of Chemical Engineering and Chemistry, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
11
|
Liu Z, Sun M, Zhang W, Ren J, Qu X. Target-Specific Bioorthogonal Reactions for Precise Biomedical Applications. Angew Chem Int Ed Engl 2023; 62:e202308396. [PMID: 37548083 DOI: 10.1002/anie.202308396] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/27/2023] [Accepted: 08/04/2023] [Indexed: 08/08/2023]
Abstract
Bioorthogonal chemistry is a promising toolbox for dissecting biological processes in the native environment. Recently, bioorthogonal reactions have attracted considerable attention in the medical field for treating diseases, since this approach may lead to improved drug efficacy and reduced side effects via in situ drug synthesis. For precise biomedical applications, it is a prerequisite that the reactions should occur in the right locations and on the appropriate therapeutic targets. In this minireview, we highlight the design and development of targeted bioorthogonal reactions for precise medical treatment. First, we compile recent strategies for achieving target-specific bioorthogonal reactions. Further, we emphasize their application for the precise treatment of different therapeutic targets. Finally, a perspective is provided on the challenges and future directions of this emerging field for safe, efficient, and translatable disease treatment.
Collapse
Affiliation(s)
- Zhengwei Liu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
| | - Mengyu Sun
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Wenting Zhang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| |
Collapse
|
12
|
Liu X, Huang T, Chen Z, Yang H. Progress in controllable bioorthogonal catalysis for prodrug activation. Chem Commun (Camb) 2023; 59:12548-12559. [PMID: 37791560 DOI: 10.1039/d3cc04286c] [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: 10/05/2023]
Abstract
Bioorthogonal catalysis, a class of catalytic reactions that are mediated by abiotic metals and proceed in biological environments without interfering with native biochemical reactions, has gained ever-increasing momentum in prodrug delivery over the past few decades. Albeit great progress has been attained in developing new bioorthogonal catalytic reactions and optimizing the catalytic performance of transition metal catalysts (TMCs), the use of TMCs to activate chemotherapeutics at the site of interest in vivo remains a challenging endeavor. To translate the bioorthogonal catalysis-mediated prodrug activation paradigm from flasks to animals, TMCs with targeting capability and stimulus-responsive behavior have been well-designed to perform chemical transformations in a controlled manner within highly complex biochemical systems, rendering on-demand drug activation to mitigate off-target toxicity. Here, we review the recent advances in the development of controllable bioorthogonal catalysis systems, with an emphasis on different strategies for engineering TMCs to achieve precise control over prodrug activation. Furthermore, we outline the envisaged challenges and discuss future directions of controllable bioorthogonal catalysis for disease therapy.
Collapse
Affiliation(s)
- Xia Liu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, and Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou 350108, P. R. China.
| | - Tingjing Huang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, and Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou 350108, P. R. China.
| | - Zhaowei Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, and Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou 350108, P. R. China.
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China.
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, and Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou 350108, P. R. China.
| |
Collapse
|
13
|
Nasibullin I, Yoshioka H, Mukaimine A, Nakamura A, Kusakari Y, Chang TC, Tanaka K. Catalytic olefin metathesis in blood. Chem Sci 2023; 14:11033-11039. [PMID: 37860663 PMCID: PMC10583672 DOI: 10.1039/d3sc03785a] [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: 07/22/2023] [Accepted: 09/05/2023] [Indexed: 10/21/2023] Open
Abstract
The direct synthesis of drugs in vivo enables drugs to treat diseases without causing side effects in healthy tissues. Transition-metal reactions have been widely explored for uncaging and synthesizing bioactive drugs in biological environments because of their remarkable reactivity. Nonetheless, it is difficult to develop a promising method to achieve in vivo drug synthesis because blood cells and metabolites deactivate transition-metal catalysts. We report that a robust albumin-based artificial metalloenzyme (ArM) with a low loading (1-5 mol%) can promote Ru-based olefin metathesis to synthesize molecular scaffolds and an antitumor drug in blood. The ArM retained its activity after soaking in blood for 24 h and provided the first example of catalytic olefin cross metathesis in blood. Furthermore, the cyclic-Arg-Gly-Asp (cRGD) peptide-functionalized ArM at lower dosages could still efficiently perform in vivo drug synthesis to inhibit the growth of implanted tumors in mice. Such a system can potentially construct therapeutic drugs in vivo for therapies without side effects.
Collapse
Affiliation(s)
- Igor Nasibullin
- Biofunctional Synthetic Chemistry Laboratory, Cluster for Pioneering Research RIKEN Wako-shi Saitama 351-0198 Japan
| | - Hiromasa Yoshioka
- Biofunctional Synthetic Chemistry Laboratory, Cluster for Pioneering Research RIKEN Wako-shi Saitama 351-0198 Japan
| | - Akari Mukaimine
- Biofunctional Synthetic Chemistry Laboratory, Cluster for Pioneering Research RIKEN Wako-shi Saitama 351-0198 Japan
| | - Akiko Nakamura
- Biofunctional Synthetic Chemistry Laboratory, Cluster for Pioneering Research RIKEN Wako-shi Saitama 351-0198 Japan
| | - Yuriko Kusakari
- Biofunctional Synthetic Chemistry Laboratory, Cluster for Pioneering Research RIKEN Wako-shi Saitama 351-0198 Japan
| | - Tsung-Che Chang
- Biofunctional Synthetic Chemistry Laboratory, Cluster for Pioneering Research RIKEN Wako-shi Saitama 351-0198 Japan
| | - Katsunori Tanaka
- Biofunctional Synthetic Chemistry Laboratory, Cluster for Pioneering Research RIKEN Wako-shi Saitama 351-0198 Japan
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology Meguro-ku Tokyo 152-8552 Japan
| |
Collapse
|
14
|
Śmiłowicz D, Eisenberg S, LaForest R, Whetter J, Hariharan A, Bordenca J, Johnson CJ, Boros E. Metal-Mediated, Autolytic Amide Bond Cleavage: A Strategy for the Selective, Metal Complexation-Catalyzed, Controlled Release of Metallodrugs. J Am Chem Soc 2023; 145:16261-16270. [PMID: 37434328 PMCID: PMC10530410 DOI: 10.1021/jacs.3c05492] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Activation of metalloprodrugs or prodrug activation using transition metal catalysts represents emerging strategies for drug development; however, they are frequently hampered by poor spatiotemporal control and limited catalytic turnover. Here, we demonstrate that metal complex-mediated, autolytic release of active metallodrugs can be successfully employed to prepare clinical grade (radio-)pharmaceuticals. Optimization of the Lewis-acidic metal ion, chelate, amino acid linker, and biological targeting vector provides means to release peptide-based (radio-)metallopharmaceuticals in solution and from the solid phase using metal-mediated, autolytic amide bond cleavage (MMAAC). Our findings indicate that coordinative polarization of an amide bond by strong, trivalent Lewis acids such as Ga3+ and Sc3+ adjacent to serine results in the N, O acyl shift and hydrolysis of the corresponding ester without dissociation of the corresponding metal complex. Compound [68Ga]Ga-10, incorporating a cleavable and noncleavable functionalization, was used to demonstrate that only the amide bond-adjacent serine effectively triggered hydrolysis in solution and from the solid phase. The corresponding solid-phase released compound [68Ga]Ga-8 demonstrated superior in vivo performance in a mouse tumor model compared to [68Ga]Ga-8 produced using conventional, solution-phase radiolabeling. A second proof-of-concept system, [67Ga]Ga-17A (serine-linked) and [67Ga]Ga-17B (glycine-linked) binding to serum albumin via the incorporated ibuprofen moiety, was also synthesized. These constructs demonstrated that complete hydrolysis of the corresponding [68Ga]Ga-NOTA complex from [67Ga]Ga-17A can be achieved in naïve mice within 12 h, as traceable in urine and blood metabolites. The glycine-linked control [68Ga]Ga-17B remained intact. Conclusively, MMAAC provides an attractive tool for selective, thermal, and metal ion-mediated control of metallodrug activation compatible with biological conditions.
Collapse
Affiliation(s)
- Dariusz Śmiłowicz
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - Shawn Eisenberg
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
| | - Rochelle LaForest
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
| | - Jennifer Whetter
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - Annapoorani Hariharan
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
| | - Jake Bordenca
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
| | - Christopher J Johnson
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
| | - Eszter Boros
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| |
Collapse
|
15
|
Dal Forno GM, Latocheski E, Beatriz Machado A, Becher J, Dunsmore L, St John AL, Oliveira BL, Navo CD, Jiménez-Osés G, Fior R, Domingos JB, Bernardes GJL. Expanding Transition Metal-Mediated Bioorthogonal Decaging to Include C-C Bond Cleavage Reactions. J Am Chem Soc 2023; 145:10790-10799. [PMID: 37133984 DOI: 10.1021/jacs.3c01960] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The ability to control the activation of prodrugs by transition metals has been shown to have great potential for controlled drug release in cancer cells. However, the strategies developed so far promote the cleavage of C-O or C-N bonds, which limits the scope of drugs to only those that present amino or hydroxyl groups. Here, we report the decaging of an ortho-quinone prodrug, a propargylated β-lapachone derivative, through a palladium-mediated C-C bond cleavage. The reaction's kinetic and mechanistic behavior was studied under biological conditions along with computer modeling. The results indicate that palladium (II) is the active species for the depropargylation reaction, activating the triple bond for nucleophilic attack by a water molecule before the C-C bond cleavage takes place. Palladium iodide nanoparticles were found to efficiently trigger the C-C bond cleavage reaction under biocompatible conditions. In drug activation assays in cells, the protected analogue of β-lapachone was activated by nontoxic amounts of nanoparticles, which restored drug toxicity. The palladium-mediated ortho-quinone prodrug activation was further demonstrated in zebrafish tumor xenografts, which resulted in a significant anti-tumoral effect. This work expands the transition-metal-mediated bioorthogonal decaging toolbox to include cleavage of C-C bonds and payloads that were previously not accessible by conventional strategies.
Collapse
Affiliation(s)
- Gean M Dal Forno
- Department of Chemistry, Federal University of Santa Catarina─UFSC, Campus Trindade, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Eloah Latocheski
- Department of Chemistry, Federal University of Santa Catarina─UFSC, Campus Trindade, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Ana Beatriz Machado
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Av. Brasilia, Lisboa 1400-038, Portugal
| | - Julie Becher
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| | - Lavinia Dunsmore
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| | - Albert L St John
- Department of Chemistry, Federal University of Santa Catarina─UFSC, Campus Trindade, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Bruno L Oliveira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, Lisboa 1649-028, Portugal
| | - 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
| | - 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, Bilbao 48013, Spain
| | - Rita Fior
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Av. Brasilia, Lisboa 1400-038, Portugal
| | - Josiel B Domingos
- Department of Chemistry, Federal University of Santa Catarina─UFSC, Campus Trindade, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Gonçalo J L Bernardes
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, Lisboa 1649-028, Portugal
| |
Collapse
|
16
|
Fedeli S, Huang R, Oz Y, Zhang X, Gupta A, Gopalakrishnan S, Makabenta JMV, Lamkin S, Sanyal A, Xu Y, Rotello VM. Biodegradable Antibacterial Bioorthogonal Polymeric Nanocatalysts Prepared by Flash Nanoprecipitation. ACS APPLIED MATERIALS & INTERFACES 2023; 15:15260-15268. [PMID: 36920076 PMCID: PMC10699753 DOI: 10.1021/acsami.3c02640] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Bioorthogonal activation of pro-dyes and prodrugs using transition-metal catalysts (TMCs) provides a promising strategy for imaging and therapeutic applications. TMCs can be loaded into polymeric nanoparticles through hydrophobic encapsulation to generate polymeric nanocatalysts with enhanced solubility and stability. However, biomedical use of these nanostructures faces challenges due to unwanted tissue accumulation of nonbiodegradable nanomaterials and cytotoxicity of heavy-metal catalysts. We report here the creation of fully biodegradable nanocatalysts based on an engineered FDA-approved polymer and the naturally existing catalyst hemin. Stable nanocatalysts were generated through kinetic stabilization using flash nanoprecipitation. The therapeutic potential of these nanocatalysts was demonstrated through effective treatment of bacterial biofilms through the bioorthogonal activation of a pro-antibiotic.
Collapse
Affiliation(s)
- Stefano Fedeli
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Rui Huang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Yavuz Oz
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
- Department of Chemistry, Bogazici University, Istanbul 34342, Turkey
| | - 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
| | - Sanjana Gopalakrishnan
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Jessa Marie V. Makabenta
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Stephanie Lamkin
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Amitav Sanyal
- Department of Chemistry, Bogazici University, Istanbul 34342, Turkey
| | - 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
|
17
|
Pérez-López AM, Belsom A, Fiedler L, Xin X, Rappsilber J. Dual-Bioorthogonal Catalysis by a Palladium Peptide Complex. J Med Chem 2023; 66:3301-3311. [PMID: 36820649 PMCID: PMC10009749 DOI: 10.1021/acs.jmedchem.2c01689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Artificial metalloenzymes (ArMs) enrich bioorthogonal chemistry with new-to-nature reactions while limiting metal deactivation and toxicity. This enables biomedical applications such as activating therapeutics in situ. However, while combination therapies are becoming widespread anticancer treatments, dual catalysis by ArMs has not yet been shown. We present a heptapeptidic ArM with a novel peptide ligand carrying a methyl salicylate palladium complex. We observed that the peptide scaffold reduces metal toxicity while protecting the metal from deactivation by cellular components. Importantly, the peptide also improves catalysis, suggesting involvement in the catalytic reaction mechanism. Our work shows how a palladium-peptide homogeneous catalyst can simultaneously mediate two types of chemistry to synthesize anticancer drugs in human cells. Methyl salicylate palladium LLEYLKR peptide (2-Pd) succeeded to simultaneously produce paclitaxel by depropargylation, and linifanib by Suzuki-Miyaura cross-coupling in cell culture, thereby achieving combination therapy on non-small-cell lung cancer (NSCLC) A549 cells.
Collapse
Affiliation(s)
- Ana M Pérez-López
- Chair of Bioanalytics, Technische Universität Berlin, 10623 Berlin, Germany.,Si-M/"Der Simulierte Mensch", a Science Framework of Technische Universität Berlin and Charité─Universitätsmedizin Berlin, 10623 Berlin, Germany
| | - Adam Belsom
- Chair of Bioanalytics, Technische Universität Berlin, 10623 Berlin, Germany.,Si-M/"Der Simulierte Mensch", a Science Framework of Technische Universität Berlin and Charité─Universitätsmedizin Berlin, 10623 Berlin, Germany
| | - Linus Fiedler
- Chair of Bioanalytics, Technische Universität Berlin, 10623 Berlin, Germany.,Si-M/"Der Simulierte Mensch", a Science Framework of Technische Universität Berlin and Charité─Universitätsmedizin Berlin, 10623 Berlin, Germany
| | - Xiaoyi Xin
- Chair of Bioanalytics, Technische Universität Berlin, 10623 Berlin, Germany.,Si-M/"Der Simulierte Mensch", a Science Framework of Technische Universität Berlin and Charité─Universitätsmedizin Berlin, 10623 Berlin, Germany
| | - Juri Rappsilber
- Chair of Bioanalytics, Technische Universität Berlin, 10623 Berlin, Germany.,Si-M/"Der Simulierte Mensch", a Science Framework of Technische Universität Berlin and Charité─Universitätsmedizin Berlin, 10623 Berlin, Germany.,Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, U.K
| |
Collapse
|
18
|
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]
|
19
|
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: 16] [Impact Index Per Article: 8.0] [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
|
20
|
Zhang L, Sang Y, Liu Z, Wang W, Liu Z, Deng Q, You Y, Ren J, Qu X. Liquid Metal as Bioinspired and Unusual Modulator in Bioorthogonal Catalysis for Tumor Inhibition Therapy. Angew Chem Int Ed Engl 2023; 62:e202218159. [PMID: 36578232 DOI: 10.1002/anie.202218159] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 12/30/2022]
Abstract
Bioorthogonal catalysis mediated by Pd-based transition metal catalysts has sparked increasing interest in combating diseases. However, the catalytic and therapeutic efficiency of current Pd0 catalysts is unsatisfactory. Herein, inspired by the concept that ligands around metal sites could enable enzymes to catalyze astonishing reactions by changing their electronic environment, a LM-Pd catalyst with liquid metal (LM) as an unusual modulator has been designed to realize efficient bioorthogonal catalysis for tumor inhibition. The LM matrix can serve as a "ligand" to afford an electron-rich environment to stabilize the active Pd0 and promote nucleophilic turnover of the π-allylpalladium species to accelerate the uncaging process. Besides, the photothermal properties of LM can lead to the enhanced removal of tumor cells by photo-enhanced catalysis and photothermal effect. We believe that our work will broaden the application of LM and motivate the design of bioinspired bioorthogonal catalysts.
Collapse
Affiliation(s)
- Lu Zhang
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, 130022, Changchun, Jilin, P. R. China.,University of Chinese Academy of Sciences, 100039, Beijing, China
| | - Yanjuan Sang
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, 130022, Changchun, Jilin, P. R. China
| | - Zhenqi Liu
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, 130022, Changchun, Jilin, P. R. China.,University of Science and Technology of China, 230026, Hefei, Anhui, China
| | - Wenjie Wang
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, 130022, Changchun, Jilin, P. R. China.,University of Science and Technology of China, 230026, Hefei, Anhui, China
| | - Zhengwei Liu
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, 130022, Changchun, Jilin, P. R. China.,University of Chinese Academy of Sciences, 100039, Beijing, China
| | - Qingqing Deng
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, 130022, Changchun, Jilin, P. R. China.,University of Science and Technology of China, 230026, Hefei, Anhui, China
| | - Yawen You
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, 130022, Changchun, Jilin, P. R. China.,University of Science and Technology of China, 230026, Hefei, Anhui, China
| | - Jinsong Ren
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, 130022, Changchun, Jilin, P. R. China.,University of Chinese Academy of Sciences, 100039, Beijing, China.,University of Science and Technology of China, 230026, Hefei, Anhui, China
| | - Xiaogang Qu
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, 130022, Changchun, Jilin, P. R. China.,University of Chinese Academy of Sciences, 100039, Beijing, China.,University of Science and Technology of China, 230026, Hefei, Anhui, China
| |
Collapse
|
21
|
Rubio-Ruiz B, Pérez-López AM, Uson L, Ortega-Liebana MC, Valero T, Arruebo M, Hueso JL, Sebastian V, Santamaria J, Unciti-Broceta A. In Cellulo Bioorthogonal Catalysis by Encapsulated AuPd Nanoalloys: Overcoming Intracellular Deactivation. NANO LETTERS 2023; 23:804-811. [PMID: 36648322 PMCID: PMC9912372 DOI: 10.1021/acs.nanolett.2c03593] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/09/2023] [Indexed: 06/17/2023]
Abstract
Bioorthogonal metallocatalysis has opened up a xenobiotic route to perform nonenzymatic catalytic transformations in living settings. Despite their promising features, most metals are deactivated inside cells by a myriad of reactive biomolecules, including biogenic thiols, thereby limiting the catalytic functioning of these abiotic reagents. Here we report the development of cytocompatible alloyed AuPd nanoparticles with the capacity to elicit bioorthogonal depropargylations with high efficiency in biological media. We also show that the intracellular catalytic performance of these nanoalloys is significantly enhanced by protecting them following two different encapsulation methods. Encapsulation in mesoporous silica nanorods resulted in augmented catalyst reactivity, whereas the use of a biodegradable PLGA matrix increased nanoalloy delivery across the cell membrane. The functional potential of encapsulated AuPd was demonstrated by releasing the potent chemotherapy drug paclitaxel inside cancer cells. Nanoalloy encapsulation provides a novel methodology to develop nanoreactors capable of mediating new-to-life reactions in cells.
Collapse
Affiliation(s)
- Belén Rubio-Ruiz
- Edinburgh
Cancer Research, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, U.K.
- Department
of Medicinal and Organic Chemistry and Unit of Excellence in Chemistry
Applied to Biomedicine and Environment, Faculty of Pharmacy, Campus
Cartuja s/n, University of Granada, 18071 Granada, Spain
- GENYO,
Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avda. Ilustración 114, 18016 Granada, Spain
| | - Ana M. Pérez-López
- Edinburgh
Cancer Research, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, U.K.
- TU
Berlin, Institut für
Biotechnologie, Aufgang
17-1, Level 4, Raum 472, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Laura Uson
- Instituto
de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain
- Department
of Chemical Engineering and Environmental Technologies, University of Zaragoza, 50018 Zaragoza, Spain
| | - M. Carmen Ortega-Liebana
- Edinburgh
Cancer Research, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, U.K.
- Department
of Medicinal and Organic Chemistry and Unit of Excellence in Chemistry
Applied to Biomedicine and Environment, Faculty of Pharmacy, Campus
Cartuja s/n, University of Granada, 18071 Granada, Spain
- GENYO,
Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avda. Ilustración 114, 18016 Granada, Spain
| | - Teresa Valero
- Edinburgh
Cancer Research, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, U.K.
- Department
of Medicinal and Organic Chemistry and Unit of Excellence in Chemistry
Applied to Biomedicine and Environment, Faculty of Pharmacy, Campus
Cartuja s/n, University of Granada, 18071 Granada, Spain
- GENYO,
Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avda. Ilustración 114, 18016 Granada, Spain
| | - Manuel Arruebo
- Instituto
de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain
- Department
of Chemical Engineering and Environmental Technologies, University of Zaragoza, 50018 Zaragoza, Spain
- Networking
Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-
BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jose L. Hueso
- Instituto
de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain
- Department
of Chemical Engineering and Environmental Technologies, University of Zaragoza, 50018 Zaragoza, Spain
- Networking
Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-
BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Victor Sebastian
- Instituto
de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain
- Department
of Chemical Engineering and Environmental Technologies, University of Zaragoza, 50018 Zaragoza, Spain
- Networking
Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-
BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jesus Santamaria
- Instituto
de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain
- Department
of Chemical Engineering and Environmental Technologies, University of Zaragoza, 50018 Zaragoza, Spain
- Networking
Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-
BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Asier Unciti-Broceta
- Edinburgh
Cancer Research, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, U.K.
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
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
|
25
|
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
|
26
|
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
|
27
|
Ortega‐Liebana MC, Porter NJ, Adam C, Valero T, Hamilton L, Sieger D, Becker CG, Unciti‐Broceta A. Truly-Biocompatible Gold Catalysis Enables Vivo-Orthogonal Intra-CNS Release of Anxiolytics. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 134:e202111461. [PMID: 38505566 PMCID: PMC10946786 DOI: 10.1002/ange.202111461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/23/2021] [Indexed: 03/21/2024]
Abstract
Being recognized as the best-tolerated of all metals, the catalytic potential of gold (Au) has thus far been hindered by the ubiquitous presence of thiols in organisms. Herein we report the development of a truly-catalytic Au-polymer composite by assembling ultrasmall Au-nanoparticles at the protein-repelling outer layer of a co-polymer scaffold via electrostatic loading. Illustrating the in vivo-compatibility of the novel catalysts, we show their capacity to uncage the anxiolytic agent fluoxetine at the central nervous system (CNS) of developing zebrafish, influencing their swim pattern. This bioorthogonal strategy has enabled -for the first time- modification of cognitive activity by releasing a neuroactive agent directly in the brain of an animal.
Collapse
Affiliation(s)
- M. Carmen Ortega‐Liebana
- Cancer Research UK Edinburgh CentreInstitute of Genetics & CancerUniversity of EdinburghEdinburghEH4 2XUUK
| | - Nicola J. Porter
- Centre for Discovery Brain SciencesThe Chancellor's BuildingUniversity of EdinburghEdinburghEH16 4SBUK
| | - Catherine Adam
- Cancer Research UK Edinburgh CentreInstitute of Genetics & CancerUniversity of EdinburghEdinburghEH4 2XUUK
| | - Teresa Valero
- Cancer Research UK Edinburgh CentreInstitute of Genetics & CancerUniversity of EdinburghEdinburghEH4 2XUUK
| | - Lloyd Hamilton
- Centre for Discovery Brain SciencesThe Chancellor's BuildingUniversity of EdinburghEdinburghEH16 4SBUK
| | - Dirk Sieger
- Centre for Discovery Brain SciencesThe Chancellor's BuildingUniversity of EdinburghEdinburghEH16 4SBUK
| | - Catherina G. Becker
- Centre for Discovery Brain SciencesThe Chancellor's BuildingUniversity of EdinburghEdinburghEH16 4SBUK
- Center for Regenerative TherapiesTechnische Universität Dresden01307DresdenGermany
| | - Asier Unciti‐Broceta
- Cancer Research UK Edinburgh CentreInstitute of Genetics & CancerUniversity of EdinburghEdinburghEH4 2XUUK
| |
Collapse
|
28
|
Ortega‐Liebana MC, Porter NJ, Adam C, Valero T, Hamilton L, Sieger D, Becker CG, Unciti‐Broceta A. Truly-Biocompatible Gold Catalysis Enables Vivo-Orthogonal Intra-CNS Release of Anxiolytics. Angew Chem Int Ed Engl 2022; 61:e202111461. [PMID: 34730266 PMCID: PMC9299494 DOI: 10.1002/anie.202111461] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/23/2021] [Indexed: 01/07/2023]
Abstract
Being recognized as the best-tolerated of all metals, the catalytic potential of gold (Au) has thus far been hindered by the ubiquitous presence of thiols in organisms. Herein we report the development of a truly-catalytic Au-polymer composite by assembling ultrasmall Au-nanoparticles at the protein-repelling outer layer of a co-polymer scaffold via electrostatic loading. Illustrating the in vivo-compatibility of the novel catalysts, we show their capacity to uncage the anxiolytic agent fluoxetine at the central nervous system (CNS) of developing zebrafish, influencing their swim pattern. This bioorthogonal strategy has enabled -for the first time- modification of cognitive activity by releasing a neuroactive agent directly in the brain of an animal.
Collapse
Affiliation(s)
- M. Carmen Ortega‐Liebana
- Cancer Research UK Edinburgh CentreInstitute of Genetics & CancerUniversity of EdinburghEdinburghEH4 2XUUK
| | - Nicola J. Porter
- Centre for Discovery Brain SciencesThe Chancellor's BuildingUniversity of EdinburghEdinburghEH16 4SBUK
| | - Catherine Adam
- Cancer Research UK Edinburgh CentreInstitute of Genetics & CancerUniversity of EdinburghEdinburghEH4 2XUUK
| | - Teresa Valero
- Cancer Research UK Edinburgh CentreInstitute of Genetics & CancerUniversity of EdinburghEdinburghEH4 2XUUK
| | - Lloyd Hamilton
- Centre for Discovery Brain SciencesThe Chancellor's BuildingUniversity of EdinburghEdinburghEH16 4SBUK
| | - Dirk Sieger
- Centre for Discovery Brain SciencesThe Chancellor's BuildingUniversity of EdinburghEdinburghEH16 4SBUK
| | - Catherina G. Becker
- Centre for Discovery Brain SciencesThe Chancellor's BuildingUniversity of EdinburghEdinburghEH16 4SBUK
- Center for Regenerative TherapiesTechnische Universität Dresden01307DresdenGermany
| | - Asier Unciti‐Broceta
- Cancer Research UK Edinburgh CentreInstitute of Genetics & CancerUniversity of EdinburghEdinburghEH4 2XUUK
| |
Collapse
|
29
|
Velasco-Lozano S, Castro SAD, Sanchez-Cano C, Benítez-Mateos AI, López-Gallego F, Salassa L. Metal substrate catalysis in the confined space for platinum drug delivery. Chem Sci 2021; 13:59-67. [PMID: 35059151 PMCID: PMC8694326 DOI: 10.1039/d1sc05151b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/22/2021] [Indexed: 01/10/2023] Open
Abstract
Catalysis-based approaches for the activation of anticancer agents hold considerable promise. These principally rely on the use of metal catalysts capable of deprotecting inactive precursors of organic drugs or transforming key biomolecules available in the cellular environment. Nevertheless, the efficiency of most of the schemes described so far is rather low, limiting the benefits of catalytic amplification as strategy for controlling the therapeutic effects of anticancer compounds. In the work presented here, we show that flavin reactivity within a hydrogel matrix provides a viable solution for the efficient catalytic activation and delivery of cisplatin, a worldwide clinically-approved inorganic chemotherapy agent. This is achieved by ionically adsorbing a flavin catalyst and a Pt(iv) prodrug as substrate into porous amino-functionalized agarose beads. The hydrogel chassis supplies high local concentrations of electron donating groups/molecules in the surrounding of the catalyst, ultimately boosting substrate conversion rates (TOF >200 min-1) and enabling controlled liberation of the drug by light or chemical stimuli. Overall, this approach can afford platforms for the efficient delivery of platinum drugs as demonstrated herein by using a transdermal diffusion model simulating the human skin.
Collapse
Affiliation(s)
- Susana Velasco-Lozano
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA) Paseo de Miramon 182 San Sebastián 20014 Spain
| | | | - Carlos Sanchez-Cano
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA) Paseo de Miramon 182 San Sebastián 20014 Spain
- Donostia International Physics Center Paseo Manuel de Lardizabal 4 Donostia 20018 Spain
- Ikerbasque, Basque Foundation for Science Bilbao 48011 Spain
| | - Ana I Benítez-Mateos
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA) Paseo de Miramon 182 San Sebastián 20014 Spain
| | - Fernando López-Gallego
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA) Paseo de Miramon 182 San Sebastián 20014 Spain
- Ikerbasque, Basque Foundation for Science Bilbao 48011 Spain
| | - Luca Salassa
- Donostia International Physics Center Paseo Manuel de Lardizabal 4 Donostia 20018 Spain
- Ikerbasque, Basque Foundation for Science Bilbao 48011 Spain
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, Euskal Herriko Unibertsitatea UPV/EHU Paseo Manuel de Lardizabal 3 Donostia 20018 Spain
| |
Collapse
|
30
|
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: 66] [Impact Index Per Article: 16.5] [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
|
31
|
Chen X, Xu T, Zhu X, Chen L, Yang L, Wang S, Yu Z, Wang L, Zhang J, Zhou H. Lamellar Metal Oxide Based Nanoagent Realizing Intensive Interlamellar Ca 2+ Release and Hypoxia Relief for Enhanced Synergistic Therapy. ACS APPLIED BIO MATERIALS 2021; 4:7993-8003. [DOI: 10.1021/acsabm.1c00894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Xiaoqin Chen
- College of Chemistry and Chemical Engineering, Anhui University, No. 111 Jiulong Road, Hefei 230601, P. R. China
| | - Tianren Xu
- College of Chemistry and Chemical Engineering, Anhui University, No. 111 Jiulong Road, Hefei 230601, P. R. China
| | - Xiaojiao Zhu
- College of Chemistry and Chemical Engineering, Anhui University, No. 111 Jiulong Road, Hefei 230601, P. R. China
| | - Lei Chen
- College of Chemistry and Chemical Engineering, Anhui University, No. 111 Jiulong Road, Hefei 230601, P. R. China
| | - Li Yang
- College of Chemistry and Chemical Engineering, Anhui University, No. 111 Jiulong Road, Hefei 230601, P. R. China
| | - Shengnan Wang
- College of Chemistry and Chemical Engineering, Anhui University, No. 111 Jiulong Road, Hefei 230601, P. R. China
| | - Zhipeng Yu
- Institute of Physical Science and Information Technology, Anhui University, Hefei 230601, P. R. China
| | - Lianke Wang
- Institute of Physical Science and Information Technology, Anhui University, Hefei 230601, P. R. China
| | - Jie Zhang
- Institute of Physical Science and Information Technology, Anhui University, Hefei 230601, P. R. China
| | - Hongping Zhou
- College of Chemistry and Chemical Engineering, Anhui University, No. 111 Jiulong Road, Hefei 230601, P. R. China
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
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
|
35
|
Chen Z, Li H, Bian Y, Wang Z, Chen G, Zhang X, Miao Y, Wen D, Wang J, Wan G, Zeng Y, Abdou P, Fang J, Li S, Sun CJ, Gu Z. Bioorthogonal catalytic patch. NATURE NANOTECHNOLOGY 2021; 16:933-941. [PMID: 33972760 DOI: 10.1038/s41565-021-00910-7] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 03/29/2021] [Indexed: 05/23/2023]
Abstract
Bioorthogonal catalysis mediated by transition metals has inspired a new subfield of artificial chemistry complementary to enzymatic reactions, enabling the selective labelling of biomolecules or in situ synthesis of bioactive agents via non-natural processes. However, the effective deployment of bioorthogonal catalysis in vivo remains challenging, mired by the safety concerns of metal toxicity or complicated procedures to administer catalysts. Here, we describe a bioorthogonal catalytic device comprising a microneedle array patch integrated with Pd nanoparticles deposited on TiO2 nanosheets. This device is robust and removable, and can mediate the local conversion of caged substrates into their active states in high-level living systems. In particular, we show that such a patch can promote the activation of a prodrug at subcutaneous tumour sites, restoring its parent drug's therapeutic anticancer properties. This in situ applied device potentiates local treatment efficacy and eliminates off-target prodrug activation and dose-dependent side effects in healthy organs or distant tissues.
Collapse
Affiliation(s)
- Zhaowei Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P. R. China
- Institute of Food Safety and Environment Monitoring, College of Chemistry, Fuzhou University, Fuzhou, P. R. China
- Department of Bioengineering, University of California, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, CA, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
| | - Hongjun Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P. R. China
- Department of Bioengineering, University of California, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, CA, USA
- Zhejiang Laboratory of Systems and Precision Medicine, Zhejiang University Medical Center, Hangzhou, P. R. China
| | - Yijie Bian
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, P. R. China
| | - Zejun Wang
- Department of Bioengineering, University of California, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, CA, USA
| | - Guojun Chen
- Department of Bioengineering, University of California, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, CA, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
| | - Xudong Zhang
- Department of Bioengineering, University of California, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, CA, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
| | - Yimin Miao
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, P. R. China
| | - Di Wen
- Department of Bioengineering, University of California, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, CA, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
| | - Jinqiang Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P. R. China
- Department of Bioengineering, University of California, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, CA, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
| | - Gang Wan
- Materials Science Division, Argonne National Laboratory, Lemont, IL, USA
| | - Yi Zeng
- Department of Bioengineering, University of California, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, CA, USA
| | - Peter Abdou
- Department of Bioengineering, University of California, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, CA, USA
| | - Jun Fang
- Department of Bioengineering, University of California, Los Angeles, CA, USA
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, CA, USA
- Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA, USA
| | - Cheng-Jun Sun
- Advanced Photon Source, Argonne National Laboratory, Lemont, IL, USA
| | - Zhen Gu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P. R. China.
- Department of Bioengineering, University of California, Los Angeles, CA, USA.
- California NanoSystems Institute, University of California, Los Angeles, CA, USA.
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA.
- Zhejiang Laboratory of Systems and Precision Medicine, Zhejiang University Medical Center, Hangzhou, P. R. China.
- Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA.
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, P. R. China.
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| |
Collapse
|
36
|
Lozhkin B, Ward TR. Bioorthogonal strategies for the in vivo synthesis or release of drugs. Bioorg Med Chem 2021; 45:116310. [PMID: 34365101 DOI: 10.1016/j.bmc.2021.116310] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/06/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023]
Abstract
The site-specific delivery of antitumor agents is a rapidly developing field that relies on prodrug activation and uncaging strategies. For this purpose, a wide range of homogeneous and heterogeneous biocompatible activators/catalysts have been developed to convert caged drugs with low toxicity and high stability in physiological settings into active substances in a bioorthogonal manner. The current methods allow for the site-specific delivery of activators and prodrugs to organelles, target cells, or tumors in living organisms. Here, we present an overview of the latest advances in catalytic drugs, highlighting the expanding toolbox of bioorthogonal activation strategies made possible by transition metals acting as activators or catalysts.
Collapse
Affiliation(s)
- Boris Lozhkin
- Department of Chemistry, University of Basel, BPR 1096, Mattenstrasse 24a, Biopark Rosental, 4058 Basel, Switzerland
| | - Thomas R Ward
- Department of Chemistry, University of Basel, BPR 1096, Mattenstrasse 24a, Biopark Rosental, 4058 Basel, Switzerland.
| |
Collapse
|
37
|
Shieh P, Hill MR, Zhang W, Kristufek SL, Johnson JA. Clip Chemistry: Diverse (Bio)(macro)molecular and Material Function through Breaking Covalent Bonds. Chem Rev 2021; 121:7059-7121. [PMID: 33823111 DOI: 10.1021/acs.chemrev.0c01282] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In the two decades since the introduction of the "click chemistry" concept, the toolbox of "click reactions" has continually expanded, enabling chemists, materials scientists, and biologists to rapidly and selectively build complexity for their applications of interest. Similarly, selective and efficient covalent bond breaking reactions have provided and will continue to provide transformative advances. Here, we review key examples and applications of efficient, selective covalent bond cleavage reactions, which we refer to herein as "clip reactions." The strategic application of clip reactions offers opportunities to tailor the compositions and structures of complex (bio)(macro)molecular systems with exquisite control. Working in concert, click chemistry and clip chemistry offer scientists and engineers powerful methods to address next-generation challenges across the chemical sciences.
Collapse
Affiliation(s)
- Peyton Shieh
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Megan R Hill
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Wenxu Zhang
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Samantha L Kristufek
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Jeremiah A Johnson
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
38
|
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
|
39
|
Rubio-Ruiz B, Pérez-López AM, Sebastián V, Unciti-Broceta A. A minimally-masked inactive prodrug of panobinostat that is bioorthogonally activated by gold chemistry. Bioorg Med Chem 2021; 41:116217. [PMID: 34022529 DOI: 10.1016/j.bmc.2021.116217] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/04/2021] [Accepted: 05/11/2021] [Indexed: 11/28/2022]
Abstract
The recent incorporation of Au chemistry in the bioorthogonal toolbox has opened up new opportunities to deliver biologically independent reactions in living environments. Herein we report that the O-propargylation of the hydroxamate group of the potent HDAC inhibitor panobinostat leads to a vast reduction of its anticancer properties (>500-fold). We also show that this novel prodrug is converted back into panobinostat in the presence of Au catalysts in vitro and in cell culture.
Collapse
Affiliation(s)
- Belén Rubio-Ruiz
- Cancer Research UK Edinburgh Centre, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, UK.
| | - Ana M Pérez-López
- Cancer Research UK Edinburgh Centre, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, UK
| | - Víctor Sebastián
- Department of Chemical Engineering, University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain; 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, Spain
| | - Asier Unciti-Broceta
- Cancer Research UK Edinburgh Centre, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
40
|
Plunk MA, Quintana JM, Darden CM, Lawrence MC, Naziruddin B, Kane RR. Design and Catalyzed Activation of Mycophenolic Acid Prodrugs. ACS Med Chem Lett 2021; 12:812-816. [PMID: 34055230 DOI: 10.1021/acsmedchemlett.1c00079] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/05/2021] [Indexed: 01/17/2023] Open
Abstract
Mycophenolic acid (MPA) and its morpholino ester prodrug mycophenolate mofetil (MMF) are widely used in solid organ transplantation. These drugs prevent rejection due to their potent inhibition of inosine-5'-monophosphate dehydrogenase (IMPDH), an enzyme vital for lymphocyte proliferation. As a strategy to provide localized immunosuppression in cell transplantation, four mycophenolic acid prodrugs designed to release MPA by two distinct mechanisms were synthesized and characterized. A nitrobenzyl ether prodrug was effectively converted to MPA upon exposure to bacterial nitroreductase, while a propargyl ether was converted to the active drug by immobilized Pd0 nanoparticles. In vitro, both prodrugs were inactive against IMPDH and exhibited reduced toxicity relative to the active drug, suggesting their potential for providing localized immunosuppression.
Collapse
Affiliation(s)
- Michael A. Plunk
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76706, United States
| | - Jeremy M. Quintana
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76706, United States
| | - Carly M. Darden
- Institute of Biomedical Studies, Baylor University, Waco, Texas 76706, United States
| | - Michael C. Lawrence
- Institute of Biomedical Studies, Baylor University, Waco, Texas 76706, United States
- Islet Cell Laboratory, Baylor Scott and White Research Institute, Dallas, Texas 75204, United States
| | - Bashoo Naziruddin
- Institute of Biomedical Studies, Baylor University, Waco, Texas 76706, United States
- Baylor Simmons Transplant Institute, Baylor University Medical Center, Dallas, Texas 75204, United States
| | - Robert R. Kane
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76706, United States
- Institute of Biomedical Studies, Baylor University, Waco, Texas 76706, United States
| |
Collapse
|
41
|
"One-stitch" bioorthogonal prodrug activation based on cross-linked lipoic acid nanocapsules. Biomaterials 2021; 273:120823. [PMID: 33930738 DOI: 10.1016/j.biomaterials.2021.120823] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 04/06/2021] [Accepted: 04/11/2021] [Indexed: 01/23/2023]
Abstract
Bioorthogonal prodrug activation is fascinating but suffers from staggered administration of prodrug and trigger, which would not only reduce the therapeutic effect but bring great inconvenience for clinical application. Herein, we report a new cross-linked lipoic acid nanocapsules (cLANCs) based two-component bioorthogonal nanosystem for "one-stitch" prodrug activation. Due to the reversible stability of cLANCs, the loaded prodrug and trigger cannot release in advance while can react upon arrival in the tumor tissue. Moreover, the cLANCs would be degraded into dihydrolipoic acid in tumor cells to potentiate the anticancer effect of the drug synthesized in situ. The data showed that the new bioorthogonal system held a killing effect 1.63 times higher than that of parent drug 3 against human colorectal tumor cells (HT29) and a tumor inhibitory rate 34.2% higher than that of 3 against HT29 tumor xenograft model with negligible side effects. The biodistribution study showed that the "one-stitch" prodrug activation exhibited a selective accumulation of 3 in the tumor tissue compared with free 3 group (34.2 μg vs 3.56 μg of 3/g of tissue). This two-component bioorthogonal nanosystem based on cross-linked lipoic acid nanocapsules constitutes the first example of "one-stitch" bioorthogonal prodrug activation.
Collapse
|
42
|
Chang T, Vong K, Yamamoto T, Tanaka K. Prodrug Activation by Gold Artificial Metalloenzyme‐Catalyzed Synthesis of Phenanthridinium Derivatives via Hydroamination. Angew Chem Int Ed Engl 2021; 60:12446-12454. [DOI: 10.1002/anie.202100369] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Tsung‐Che Chang
- Biofunctional Synthetic Chemistry Laboratory RIKEN Cluster for Pioneering Research, RIKEN 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
| | - Kenward Vong
- Biofunctional Synthetic Chemistry Laboratory RIKEN Cluster for Pioneering Research, RIKEN 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
- GlycoTargeting Research Laboratory RIKEN Baton Zone Program, RIKEN 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
| | - Tomoya Yamamoto
- Biofunctional Synthetic Chemistry Laboratory RIKEN Cluster for Pioneering Research, RIKEN 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
| | - Katsunori Tanaka
- Biofunctional Synthetic Chemistry Laboratory RIKEN Cluster for Pioneering Research, RIKEN 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
- GlycoTargeting Research Laboratory RIKEN Baton Zone Program, RIKEN 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 Ookayama Meguro-ku Tokyo 152-8552 Japan
- Biofunctional Chemical Laboratory, A. Butlerov Institute of Chemistry Kazan Federal University 18 Kremlyovskaya Street 420008 Kazan Russia
| |
Collapse
|
43
|
Chang T, Vong K, Yamamoto T, Tanaka K. Prodrug Activation by Gold Artificial Metalloenzyme‐Catalyzed Synthesis of Phenanthridinium Derivatives via Hydroamination. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202100369] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Tsung‐Che Chang
- Biofunctional Synthetic Chemistry Laboratory RIKEN Cluster for Pioneering Research, RIKEN 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
| | - Kenward Vong
- Biofunctional Synthetic Chemistry Laboratory RIKEN Cluster for Pioneering Research, RIKEN 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
- GlycoTargeting Research Laboratory RIKEN Baton Zone Program, RIKEN 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
| | - Tomoya Yamamoto
- Biofunctional Synthetic Chemistry Laboratory RIKEN Cluster for Pioneering Research, RIKEN 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
| | - Katsunori Tanaka
- Biofunctional Synthetic Chemistry Laboratory RIKEN Cluster for Pioneering Research, RIKEN 2-1 Hirosawa Wako-shi Saitama 351-0198 Japan
- GlycoTargeting Research Laboratory RIKEN Baton Zone Program, RIKEN 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 Ookayama Meguro-ku Tokyo 152-8552 Japan
- Biofunctional Chemical Laboratory, A. Butlerov Institute of Chemistry Kazan Federal University 18 Kremlyovskaya Street 420008 Kazan Russia
| |
Collapse
|
44
|
Destito P, Vidal C, López F, Mascareñas JL. Transition Metal‐Promoted Reactions in Aqueous Media and Biological Settings. Chemistry 2021; 27:4789-4816. [DOI: 10.1002/chem.202003927] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/27/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Paolo Destito
- Centro Singular de Investigación en Química Biolóxica e Materiais, Moleculares (CIQUS) and Departamento de Química Orgánica Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain
| | - Cristian Vidal
- Centro Singular de Investigación en Química Biolóxica e Materiais, Moleculares (CIQUS) and Departamento de Química Orgánica Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain
| | - Fernando López
- Centro Singular de Investigación en Química Biolóxica e Materiais, Moleculares (CIQUS) and Departamento de Química Orgánica Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain
- Instituto de Química Orgánica General (CSIC) Juan de la Cierva 3 28006 Madrid Spain
| | - José L. Mascareñas
- Centro Singular de Investigación en Química Biolóxica e Materiais, Moleculares (CIQUS) and Departamento de Química Orgánica Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain
| |
Collapse
|
45
|
Hu T, Yan L, Wang Z, Shen W, Liang R, Yan D, Wei M. A pH-responsive ultrathin Cu-based nanoplatform for specific photothermal and chemodynamic synergistic therapy. Chem Sci 2021; 12:2594-2603. [PMID: 34164027 PMCID: PMC8179329 DOI: 10.1039/d0sc06742c] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 12/19/2020] [Indexed: 12/14/2022] Open
Abstract
Noninvasive tumor therapy requires a new generation of bionanomaterials towards sensitive response to the unique tumor microenvironment to achieve accurate and effective treatment. Herein, we have developed a tumor therapy nanoplatform by immobilizing natural glucose oxidase (GOD) onto Cu-based layered double hydroxide (CuFe-LDH) nanosheets, which for the first time integrates acid-enhanced photothermal therapy (PTT), and pH-responsive and heat-facilitated chemodynamic therapy (CDT) simultaneously. As demonstrated by EXAFS and HRTEM, CuFe-LDH nanosheets possess a considerable number of defects caused by different acid conditions, resulting in a significantly acid-enhanced photothermal conversion efficiency (83.2% at pH 5.4 vs. 46.0% at pH 7.4). Moreover, GOD/CuFe-LDH nanosheets can convert a cascade of glucose into hydroxyl radicals (˙OH) under tumor acid conditions, which is validated by a high maximum velocity (V max = 2.00 × 10-7 M) and low Michaelis-Menten constant (K M = 12.01 mM). With the combination of PTT and CDT, the tumor tissue in vivo is almost eliminated with low-dose drug injection (1 mg kg-1). Therefore, this novel pH-responsive Cu-based nanoplatform holds great promise in tumor-specific CDT/PTT synergistic therapy.
Collapse
Affiliation(s)
- Tingting Hu
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology Beijing 100029 P. R. China
| | - Liang Yan
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology Beijing 100029 P. R. China
| | - Zhengdi Wang
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology Beijing 100029 P. R. China
| | - Weicheng Shen
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology Beijing 100029 P. R. China
| | - Ruizheng Liang
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology Beijing 100029 P. R. China
| | - Dongpeng Yan
- Beijing Key Laboratory of Energy Conversion and Storage Materials, College of Chemistry, Beijing Normal University Beijing 100875 P. R. China
| | - Min Wei
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology Beijing 100029 P. R. China
| |
Collapse
|
46
|
Brewster RC, Klemencic E, Jarvis AG. Palladium in biological media: Can the synthetic chemist's most versatile transition metal become a powerful biological tool? J Inorg Biochem 2020; 215:111317. [PMID: 33310459 DOI: 10.1016/j.jinorgbio.2020.111317] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/16/2020] [Accepted: 11/16/2020] [Indexed: 12/18/2022]
Abstract
Palladium catalysed reactions are ubiquitous in synthetic organic chemistry in both organic solvents and aqueous buffers. The broad reactivity of palladium catalysis has drawn interest as a means to conduct orthogonal transformations in biological settings. Successful examples have been shown for protein modification, in vivo drug decaging and as palladium-protein biohybrid catalysts for selective catalysis. Biological media represents a challenging environment for palladium chemistry due to the presence of a multitude of chelators, catalyst poisons and a requirement for milder reaction conditions e.g. lower temperatures. This review looks to identify successful examples of palladium-catalysed reactions in the presence of proteins or cells and analyse solutions to help to overcome the challenges of working in biological systems.
Collapse
Affiliation(s)
- Richard C Brewster
- EaStCHEM School of Chemistry, Joseph Black Building, David Brewster Rd, University of Edinburgh, Edinburgh EH9 3FJ, United Kingdom
| | - Eva Klemencic
- EaStCHEM School of Chemistry, Joseph Black Building, David Brewster Rd, University of Edinburgh, Edinburgh EH9 3FJ, United Kingdom
| | - Amanda G Jarvis
- EaStCHEM School of Chemistry, Joseph Black Building, David Brewster Rd, University of Edinburgh, Edinburgh EH9 3FJ, United Kingdom.
| |
Collapse
|
47
|
Sebastian V, Sancho-Albero M, Arruebo M, Pérez-López AM, Rubio-Ruiz B, Martin-Duque P, Unciti-Broceta A, Santamaría J. Nondestructive production of exosomes loaded with ultrathin palladium nanosheets for targeted bio-orthogonal catalysis. Nat Protoc 2020; 16:131-163. [PMID: 33247282 DOI: 10.1038/s41596-020-00406-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 08/25/2020] [Indexed: 01/20/2023]
Abstract
The use of exosomes as selective delivery vehicles of therapeutic agents, such as drugs or hyperthermia-capable nanoparticles, is being intensely investigated on account of their preferential tropism toward their parental cells. However, the methods used to introduce a therapeutic load inside exosomes often involve disruption of their membrane, which may jeopardize their targeting capabilities, attributed to their surface integrins. On the other hand, in recent years bio-orthogonal catalysis has emerged as a new tool with a myriad of potential applications in medicine. These bio-orthogonal processes, often based on Pd-catalyzed chemistry, would benefit from systems capable of delivering the catalyst to target cells. It is therefore highly attractive to combine the targeting capabilities of exosomes and the bio-orthogonal potential of Pd nanoparticles to create new therapeutic vectors. In this protocol, we provide detailed information on an efficient procedure to achieve a high load of catalytically active Pd nanosheets inside exosomes, without disrupting their membranes. The protocol involves a multistage process in which exosomes are first harvested, subjected to impregnation with a Pd salt precursor followed by a mild reduction process using gas-phase CO, which acts as both a reducing and growth-directing agent to produce the desired nanosheets. The technology is scalable, and the protocol can be conducted by any researcher having basic biology and chemistry skills in ~3 d.
Collapse
Affiliation(s)
- Victor Sebastian
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza, Spain. .,Department of Chemical Engineering and Environmental Technologies, University of Zaragoza, Zaragoza, Spain. .,Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.
| | - María Sancho-Albero
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza, Spain.,Department of Chemical Engineering and Environmental Technologies, University of Zaragoza, Zaragoza, Spain.,Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Manuel Arruebo
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza, Spain.,Department of Chemical Engineering and Environmental Technologies, University of Zaragoza, Zaragoza, Spain.,Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Ana M Pérez-López
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, UK.,Institut für Biotechnologie, Technische Universität Berlin, Berlin, Germany
| | - Belén Rubio-Ruiz
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, UK.,Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO) and Department of Medicinal and Organic Chemistry, Faculty of Pharmacy, University of Granada, Granada, Spain
| | - Pilar Martin-Duque
- Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.,Instituto Aragonés de Ciencias de la Salud-Fundación Araid/IIS Aragón, Centro de Investigaciones Biomédicas de Aragón, Universidad San Jorge, Zaragoza, Spain
| | - Asier Unciti-Broceta
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Jesús Santamaría
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza, Spain.,Department of Chemical Engineering and Environmental Technologies, University of Zaragoza, Zaragoza, Spain.,Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
48
|
van de L'Isle MON, Ortega-Liebana MC, Unciti-Broceta A. Transition metal catalysts for the bioorthogonal synthesis of bioactive agents. Curr Opin Chem Biol 2020; 61:32-42. [PMID: 33147552 DOI: 10.1016/j.cbpa.2020.10.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/01/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023]
Abstract
The incorporation of abiotic transition metal catalysis into the chemical biology space has significantly expanded the tool kit of bioorthogonal chemistries accessible for cell culture and in vivo applications. A rich variety of homogeneous and heterogeneous catalysts has shown functional compatibility with physiological conditions and biostability in complex environs, enabling their exploitation as extracellular or intracellular factories of bioactive agents. Current trends in the field are focusing on investigating new metals and sophisticated catalytic devices and toward more applied activities, such as the integration of subcellular, cell- and site-targeting capabilities or the exploration of novel biomedical applications. We present herein an overview of the latest advances in the field, highlighting the increasing role of transition metals for the controlled release of therapeutics.
Collapse
Affiliation(s)
- Melissa O N van de L'Isle
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Mari Carmen Ortega-Liebana
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, 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
|
49
|
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
|
50
|
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
|