1
|
Teng Y, Wang D, Yang Z, Wang R, Ning S, Zhang R, Yang H, Feng X, Liu J, Yang L, Tian Y. Bioorthogonal strategy-triggered In situ co-activation of aggregation-induced emission photosensitizers and chemotherapeutic prodrugs for boosting synergistic chemo-photodynamic-immunotherapy. Biomaterials 2025; 317:123092. [PMID: 39793168 DOI: 10.1016/j.biomaterials.2025.123092] [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: 11/25/2024] [Revised: 01/04/2025] [Accepted: 01/04/2025] [Indexed: 01/13/2025]
Abstract
In situ activation of prodrugs or photosensitizers is a promising strategy for specifically killing tumor cells while avoiding toxic side effects. Herein, we originally develop a bioorthogonally activatable prodrug and pro-photosensitizer system to synchronously yield an aggregation-induced emission (AIE) photosensitizer and a chemotherapeutic drug for synergistic chemo-photodynamic-immunotherapy of tumors. By employing molecular engineering strategy, we rationally design a family of tetrazine-functionalized tetraphenylene-based photosensitizers, one of which (named TzPS5) exhibits a high turn-on ratio, a NIR emission, a typical AIE character, and an excellent ROS generation efficiency upon bioorthogonal-activation. With the aid of integrin- or mitochondria-pretargeting, TzPS5 is successfully applied for highly effective PDT ablation of cancer cells both in vitro and in vivo. On this basis, tumor-targeting TzPS5 (TzPS5-cRGD) is constructed and used jointly with a bioorthogonal prodrug, DOX-TCO, and the two are mutually activated to induce cooperative and tumor-specific PDT and chemotherapy, resulting in amplified therapeutic outcomes and improved biosafeties. Moreover, this combination modality elicits robust immunogenic cell death, stimulates systemic antitumor immunity, thereby suppressing both primary and distant tumors, and blocking the pulmonary tumor metastasis. This work is expected to provide a useful guidance for the rational design of activatable phototheranostic agents, and offer a new strategy for co-activation of prodrugs/pro-photosensitizers to boost synergistic antitumor chemo-photodynamic-immunotherapy.
Collapse
Affiliation(s)
- Yu Teng
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Dianyu Wang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China
| | - Ziyu Yang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Ruxuan Wang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100007, PR China
| | - Shuyi Ning
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Rongrong Zhang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Hong Yang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Xinchi Feng
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China.
| | - Jianfeng Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China.
| | - Lijun Yang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China.
| | - Yulin Tian
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China.
| |
Collapse
|
2
|
Brejchova K, Rahm M, Benova A, Domanska V, Reyes-Gutierez P, Dzubanova M, Trubacova R, Vondrackova M, Cajka T, Tencerova M, Vrabel M, Kuda O. Uncovering mechanisms of thiazolidinediones on osteogenesis and adipogenesis using spatial fluxomics. Metabolism 2025; 166:156157. [PMID: 39947516 DOI: 10.1016/j.metabol.2025.156157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/27/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
OBJECTIVE Insulin-sensitizing drugs, despite their broad use against type 2 diabetes, can adversely affect bone health, and the mechanisms underlying these side effects remain largely unclear. Here, we investigated the different metabolic effects of a series of thiazolidinediones, including rosiglitazone, pioglitazone, and the second-generation compound MSDC-0602K, on human mesenchymal stem cells (MSCs). METHODS We developed 13C subcellular metabolomic tracer analysis measuring separate mitochondrial and cytosolic metabolite pools, lipidomic network-based isotopologue models, and bioorthogonal click chemistry, to demonstrate that MSDC-0602K differentially affected bone marrow-derived MSCs (BM-MSCs) and adipose tissue-derived MSCs (AT-MSCs). In BM-MSCs, MSDC-0602K promoted osteoblastic differentiation and suppressed adipogenesis. This effect was clearly distinct from that of the earlier drugs and that on AT-MSCs. RESULTS Fluxomic data reveal unexpected differences between this drug's effect on MSCs and provide mechanistic insight into the pharmacologic inhibition of mitochondrial pyruvate carrier 1 (MPC). Our study demonstrates that MSDC-0602K retains the capacity to inhibit MPC, akin to rosiglitazone but unlike pioglitazone, enabling the utilization of alternative metabolic pathways. Notably, MSDC-0602K exhibits a limited lipogenic potential compared to both rosiglitazone and pioglitazone, each of which employs a distinct lipogenic strategy. CONCLUSIONS These findings indicate that the new-generation drugs do not compromise bone structure, offering a safer alternative for treating insulin resistance. Moreover, these results highlight the ability of cell compartment-specific metabolite labeling by click reactions and tracer metabolomics analysis of complex lipids to discover molecular mechanisms within the intersection of carbohydrate and lipid metabolism.
Collapse
Affiliation(s)
- Kristyna Brejchova
- Laboratory of Metabolism of Bioactive Lipids, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czechia
| | - Michal Rahm
- Chemistry of Bioconjugates, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 166 10 Prague, Czechia
| | - Andrea Benova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czechia
| | - Veronika Domanska
- Laboratory of Metabolism of Bioactive Lipids, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czechia
| | - Paul Reyes-Gutierez
- Chemistry of Bioconjugates, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 166 10 Prague, Czechia
| | - Martina Dzubanova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czechia; Faculty of Science, Charles University, Albertov 6, 128 00 Prague, Czech Republic
| | - Radka Trubacova
- Laboratory of Metabolism of Bioactive Lipids, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czechia
| | - Michaela Vondrackova
- Laboratory of Metabolism of Bioactive Lipids, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czechia
| | - Tomas Cajka
- Laboratory of Translational Metabolism, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czechia
| | - Michaela Tencerova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czechia
| | - Milan Vrabel
- Chemistry of Bioconjugates, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 166 10 Prague, Czechia
| | - Ondrej Kuda
- Laboratory of Metabolism of Bioactive Lipids, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czechia.
| |
Collapse
|
3
|
Fumanal Idocin A, Specklin S, Taran F. Sydnonimines: synthesis, properties and applications in chemical biology. Chem Commun (Camb) 2025; 61:5704-5718. [PMID: 40066827 DOI: 10.1039/d5cc00535c] [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: 04/12/2025]
Abstract
Sydnonimines are intriguing compounds belonging to the mesoionic family. To date, only a limited number of research groups have studied their chemistry and use in organic synthesis, medicinal chemistry and chemical biology. This review aims at providing an overview of the synthesis and the properties of sydnonimines and the most recent developments in their use as tools for chemical biology. The recent discovery that sydnonimines can act as a dipole to undergo bioorthogonal click-and-release reactions with cycloalkynes has stimulated a renewed interest from the scientific community. Given the high potential of these mesoionics, we believe that major developments are to be expected in the field of bioorthogonal chemistry.
Collapse
Affiliation(s)
- Alfonso Fumanal Idocin
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay (BioMaps), Orsay 91401, France.
| | - Simon Specklin
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay (BioMaps), Orsay 91401, France.
| | - Frédéric Taran
- Département Médicaments et Technologies pour la Santé, CEA-DMTS-SCBM, Université Paris-Saclay, 91191 Gif-sur-Yvette, France.
| |
Collapse
|
4
|
Shen R, Zhang B, Zhao L, Chang B, Zhang F, Chen Y, Fang J. A tunable stimuli-responsive module based on an α-hydroxymethyl-α,β-unsaturated carbonyl scaffold. J Mater Chem B 2025; 13:3980-3989. [PMID: 40029635 DOI: 10.1039/d4tb02818j] [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: 03/05/2025]
Abstract
The α-hydroxymethyl-α,β-unsaturated carbonyl (HMUC) scaffold represents a valuable framework for constructing nucleophile-responsive materials. However, nucleophiles are largely limited to thiols and amines. Given the ubiquity of thiols and amines in biological systems, this limitation hinders the creation of materials that can be selectively activated by exogenous stimuli. By tuning the electron density of the double bond and assessing its reactivity with various nucleophiles, we present here the discovery of the N-ethyl-2-(hydroxymethyl)acrylamide (NEHMAA) scaffold as a versatile building block for fabricating exogenous stimuli-responsive materials. The selenol species 4-cyanobenzylselenol (from its precursor bis(4-cyanobenzyl)diselenide, Se4) effectively activates NEHMAA-decorated "caged" molecules. Furthermore, the NEHMAA unit was employed to prepare prodrugs, and Se4-dependent cytotoxicity of these prodrugs was observed in cancer cells. The orthogonal reactivity between the NEHMAA unit and Se4 enriches the existing repertoire for constructing exogenous stimuli-responsive smart materials.
Collapse
Affiliation(s)
- Ruipeng Shen
- School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing, Jiangsu 210094, China.
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Baoxin Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Lanning Zhao
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Bingbing Chang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Fang Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Yating Chen
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Jianguo Fang
- School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing, Jiangsu 210094, China.
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| |
Collapse
|
5
|
Shen T, Liu X. Unveiling the photophysical mechanistic mysteries of tetrazine-functionalized fluorogenic labels. Chem Sci 2025; 16:4595-4613. [PMID: 39906389 PMCID: PMC11789511 DOI: 10.1039/d4sc07018f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/21/2025] [Indexed: 02/06/2025] Open
Abstract
Tetrazine-based fluorogenic labels are widely utilized in medical and biological studies, exhibiting substantial fluorescence enhancement (FE) following tetrazine degradation through bio-orthogonal reactions. However, the underlying mechanisms driving this fluorogenic response remain only partially resolved, particularly regarding the diminished FE efficiency in the deep-red and near-infrared (NIR) regions. This knowledge gap has impeded efforts to optimize these labels for extended emission wavelengths and improved FE ratios. This review offers a photophysical perspective, discussing the fluorescence quenching pathways (i.e., energy flows and charge separation) that regulate the fluorogenic properties exhibited in various types of tetrazine labels. Moreover, this work examines the emerging role of intramolecular rotations in certain tetrazine-based structures and the integration of additional quencher units. The proposed alternative quenching channel offers the potential to surpass traditional wavelength constraints while achieving improved FE. By examining these photophysical mechanisms, this review aims to advance the understanding of tetrazine-functionalized fluorogenic labels and provide guiding principles for their future design and practical applications.
Collapse
Affiliation(s)
- Tianruo Shen
- Science, Mathematics and Technology Cluster, Singapore University of Technology and Design 8 Somapah Road Singapore 487372 Singapore
| | - Xiaogang Liu
- Science, Mathematics and Technology Cluster, Singapore University of Technology and Design 8 Somapah Road Singapore 487372 Singapore
| |
Collapse
|
6
|
de Roode KE, Rossin R, Robillard MS. Toward Realization of Bioorthogonal Chemistry in the Clinic. Top Curr Chem (Cham) 2025; 383:12. [PMID: 40042792 PMCID: PMC11882664 DOI: 10.1007/s41061-025-00495-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025]
Abstract
In the last decade, the use of bioorthogonal chemistry toward medical applications has increased tremendously. Besides being useful for the production of pharmaceuticals, the efficient, nontoxic reactions open possibilities for the development of therapies that rely on in vivo chemistry between two bioorthogonal components. Here we discuss the latest developments in bioorthogonal chemistry, with a focus on their use in living organisms, the translation from model systems to humans, and the challenges encountered during preclinical development. We aim to provide the reader a broad presentation of the current state of the art and demonstrate the numerous possibilities that bioorthogonal reactions have for clinical use, now and in the near future.
Collapse
Affiliation(s)
- Kim E de Roode
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED, Nijmegen, The Netherlands
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Raffaella Rossin
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED, Nijmegen, The Netherlands
| | - Marc S Robillard
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED, Nijmegen, The Netherlands.
| |
Collapse
|
7
|
Pang J, Feng S, Huang B, Zhou J, Zhan L, Long YQ. Novel Bioorthogonal Theranostic Scaffold Enables on-Target Drug Release and Real Time Monitoring In Vivo. J Med Chem 2025; 68:3824-3836. [PMID: 39887307 DOI: 10.1021/acs.jmedchem.4c02965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Bioorthogonal chemistry-based prodrug strategy features spatiotemporally controlled release of therapeutic agent and/or imaging probe. However, the integration of diagnosis and therapy into a single molecule paired with a single bioorthogonal trigger remains a challenge. In this study, we devised a novel bioorthogonal theranostic scaffold amenable to the conjugation of various targeting agent and click-to-release reaction with the bioorthogonal prodrug to enable targeted drug liberation with concomitant fluorescence emission. Such one-stone-three-birds scaffold consists of a new fluorophore phenanthrodioxine (PDO) linked with a fluorescence masking group, tetrazine (Tz) which serves as a dual switch for the activation of fluorophore and drug. Further installation of a warhead of phenylboronic acid (PBA) ensures the targeted accumulation of the resultant PBA-PDO-Tz conjugate in tumor cells, thereby achieving on-demand activation of trans-cyclooctene-caged anticancer drug Doxorubicin with real-time monitoring and on-target cytotoxicity in live cells and an A549 xenograft mouse model. The targeted single trigger-dual response scaffold holds promise for precise theranostics applications in vivo.
Collapse
Affiliation(s)
- Jing Pang
- Laboratory of Medicinal Chemical Biology, College of Pharmaceutic Sciences, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Suzhou Medical College, Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Shun Feng
- Laboratory of Medicinal Chemical Biology, College of Pharmaceutic Sciences, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Suzhou Medical College, Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Bin Huang
- Laboratory of Medicinal Chemical Biology, College of Pharmaceutic Sciences, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Suzhou Medical College, Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Jujun Zhou
- Laboratory of Medicinal Chemical Biology, College of Pharmaceutic Sciences, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Suzhou Medical College, Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Linjun Zhan
- Laboratory of Medicinal Chemical Biology, College of Pharmaceutic Sciences, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Suzhou Medical College, Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Ya-Qiu Long
- Laboratory of Medicinal Chemical Biology, College of Pharmaceutic Sciences, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Suzhou Medical College, Soochow University, 199 Renai Road, Suzhou 215123, China
| |
Collapse
|
8
|
Li J, Cheng M, Zhang H, Wang Y, Guo W, Zheng Y. A Tetrazine Amplification System for Visual Detection of Trace Analytes via Click-Release Reactions. Angew Chem Int Ed Engl 2025; 64:e202414246. [PMID: 39623886 DOI: 10.1002/anie.202414246] [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: 07/28/2024] [Revised: 12/02/2024] [Indexed: 12/17/2024]
Abstract
Achieving visual detection of analytes at ultra-low concentrations in complex mixtures remains a persistent challenge. While sophisticated techniques offer single-molecule sensitivity, practical hurdles remain, necessitating tailored signal amplification systems for direct visual detection. In this study, we develop a strategy for the visualized detection of tetrazine through a "click-release-oxidation-cycle" (CROC) cascade amplification process. We systematically describe the construction and synthesis of this system, the kinetic process of click release, the kinetics of oxidation to tetrazine and its cascade amplification effect in trace amounts of tetrazine. This system is capable of amplifying the signal of tetrazine at a concentration as low as 2 nM by 105-fold, thereby providing a clearly visible purple signal. Finally, as proof of concept, we successfully apply this method to visually detect trace β-galactosidase (β-gal) and Pd2+.
Collapse
Affiliation(s)
- Jing Li
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Mingxin Cheng
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Hongbo Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Yichen Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Weiwei Guo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 211198, P. R. China
- School of Science, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Yueqin Zheng
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 211198, P. R. China
| |
Collapse
|
9
|
Bao Y, Xu Z, Cheng K, Li X, Chen F, Yuan D, Zhang F, Che ARY, Zeng X, Zhao YD, Xia J. Staudinger Reaction-Responsive Coacervates for Cytosolic Antibody Delivery and TRIM21-Mediated Protein Degradation. J Am Chem Soc 2025; 147:3830-3839. [PMID: 39805770 PMCID: PMC11783599 DOI: 10.1021/jacs.4c17054] [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: 11/30/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
A low-molecular-weight compound whose structure strikes a fine balance between hydrophobicity and hydrophilicity may form coacervates via liquid-liquid phase separation in an aqueous solution. These coacervates may encapsulate and convoy proteins across the plasma membrane into the cell. However, releasing the cargo from the vehicle to the cytosol is challenging. Here, we address this issue by designing phase-separating coacervates, which are disassembled by the bioorthogonal Staudinger reaction. We constructed and selected triphenylphosphine-based compounds that formed phase-separated coacervates in an aqueous solution. Reacting the coacervates with azides resulted in microdroplet dissolution, so they received the name Staudinger Reaction-Responsive Coacervates, SR-Coa. SR-Coa could encapsulate proteins, including antibodies, and translocate them across the plasma membrane into the cell. Further treatment of the cell with ethyl azidoacetate induced the cargo dispersion from the puncta to the cytosolic distribution. We showcased an application of the SR-Coa/ethyl azidoacetate system in facilitating the translocation of the EGFR/antibody complex into the cell, which induced EGFR degradation via the TRIM21-dependent pathway both in vitro and in vivo. Besides the membrane protein EGFR, this system could also degrade endogenous protein EZH2. Taken together, here we report a strategy of controlling molecular coacervates by a bioorthogonal reaction in the cell for cytosolic protein delivery and demonstrate its use in promoting targeted protein degradation via the proteasome-dependent pathway.
Collapse
Affiliation(s)
- Yishu Bao
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin, Hong Kong SAR 99999, China
| | - Zhiyi Xu
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin, Hong Kong SAR 99999, China
| | - Kai Cheng
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin, Hong Kong SAR 99999, China
| | - Xiaojing Li
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin, Hong Kong SAR 99999, China
| | - Fangke Chen
- Department
of Physics, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR 99999, China
| | - Dingdong Yuan
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin, Hong Kong SAR 99999, China
| | - Fang Zhang
- Britton
Chance Center for Biomedical Photonics at Wuhan National Laboratory
for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key
Laboratory, Department of Biomedical Engineering, College of Life
Science and Technology, Huazhong University
of Science and Technology, Wuhan 430074, Hubei, China
| | - Audrey Run-Yu Che
- Department
of Natural Sciences, Pitzer and Scripps
Colleges, 925 N. Mills
Ave, Claremont, California 91711, United States
| | - Xiangze Zeng
- Department
of Physics, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR 99999, China
| | - Yuan-Di Zhao
- Britton
Chance Center for Biomedical Photonics at Wuhan National Laboratory
for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key
Laboratory, Department of Biomedical Engineering, College of Life
Science and Technology, Huazhong University
of Science and Technology, Wuhan 430074, Hubei, China
| | - Jiang Xia
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin, Hong Kong SAR 99999, China
| |
Collapse
|
10
|
Rahm M, Keppel P, Šlachtová V, Dzijak R, Dračínský M, Bellová S, Reyes-Gutiérrez PE, Štěpánová S, Raffler J, Tloušťová E, Mertlíková-Kaiserová H, Mikula H, Vrabel M. Sulfonated Hydroxyaryl-Tetrazines with Increased pK a for Accelerated Bioorthogonal Click-to-Release Reactions in Cells. Angew Chem Int Ed Engl 2025; 64:e202411713. [PMID: 39298292 DOI: 10.1002/anie.202411713] [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: 06/21/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 09/21/2024]
Abstract
Bioorthogonal reactions that enable switching molecular functions by breaking chemical bonds have gained prominence, with the tetrazine-mediated cleavage of trans-cyclooctene caged compounds (click-to-release) being particularly noteworthy for its high versatility, biocompatibility, and fast reaction rates. Despite several recent advances, the development of highly reactive tetrazines enabling quantitative elimination from trans-cyclooctene linkers remains challenging. In this study, we present the synthesis and application of sulfo-tetrazines, a class of derivatives featuring phenolic hydroxyl groups with increased acidity constants (pKa). This unique property leads to accelerated elimination and complete release of the caged molecules within minutes. Moreover, the inclusion of sulfonate groups provides a valuable synthetic handle, enabling further derivatization into sulfonamides, modified with diverse substituents. Significantly, we demonstrate the utility of sulfo-tetrazines in efficiently activating fluorogenic compounds and prodrugs in living cells, offering exciting prospects for their application in bioorthogonal chemistry.
Collapse
Affiliation(s)
- Michal Rahm
- Institute of Organic Chemistry and Biochemistry of the CAS, Flemingovo nám. 2, 16000, Prague 6, Czech Republic
- University of Chemistry and Technology, Department of Chemistry of Natural Compounds, Technická 5, 166 28, Prague 6, Czech Republic
| | - Patrick Keppel
- Institute of Applied Synthetic Chemistry, TU Wien, Getreidemarkt 9, 1060, Vienna, Austria
| | - Veronika Šlachtová
- Institute of Organic Chemistry and Biochemistry of the CAS, Flemingovo nám. 2, 16000, Prague 6, Czech Republic
| | - Rastislav Dzijak
- Institute of Organic Chemistry and Biochemistry of the CAS, Flemingovo nám. 2, 16000, Prague 6, Czech Republic
| | - Martin Dračínský
- Institute of Organic Chemistry and Biochemistry of the CAS, Flemingovo nám. 2, 16000, Prague 6, Czech Republic
| | - Simona Bellová
- Institute of Organic Chemistry and Biochemistry of the CAS, Flemingovo nám. 2, 16000, Prague 6, Czech Republic
| | - Paul E Reyes-Gutiérrez
- Institute of Organic Chemistry and Biochemistry of the CAS, Flemingovo nám. 2, 16000, Prague 6, Czech Republic
| | - Sille Štěpánová
- Institute of Organic Chemistry and Biochemistry of the CAS, Flemingovo nám. 2, 16000, Prague 6, Czech Republic
| | - Jakob Raffler
- Institute of Applied Synthetic Chemistry, TU Wien, Getreidemarkt 9, 1060, Vienna, Austria
| | - Eva Tloušťová
- Institute of Organic Chemistry and Biochemistry of the CAS, Flemingovo nám. 2, 16000, Prague 6, Czech Republic
| | - Helena Mertlíková-Kaiserová
- Institute of Organic Chemistry and Biochemistry of the CAS, Flemingovo nám. 2, 16000, Prague 6, Czech Republic
| | - Hannes Mikula
- Institute of Applied Synthetic Chemistry, TU Wien, Getreidemarkt 9, 1060, Vienna, Austria
| | - Milan Vrabel
- Institute of Organic Chemistry and Biochemistry of the CAS, Flemingovo nám. 2, 16000, Prague 6, Czech Republic
| |
Collapse
|
11
|
Wilkovitsch M, Kuba W, Keppel P, Sohr B, Löffler A, Kronister S, Del Castillo AF, Goldeck M, Dzijak R, Rahm M, Vrabel M, Svatunek D, Carlson JCT, Mikula H. Transforming Aryl-Tetrazines into Bioorthogonal Scissors for Systematic Cleavage of trans-Cyclooctenes. Angew Chem Int Ed Engl 2025; 64:e202411707. [PMID: 39254137 DOI: 10.1002/anie.202411707] [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: 06/21/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024]
Abstract
Bioorthogonal bond-cleavage reactions have emerged as a powerful tool for precise spatiotemporal control of (bio)molecular function in the biological context. Among these chemistries, the tetrazine-triggered elimination of cleavable trans-cyclooctenes (click-to-release) stands out due to high reaction rates, versatility, and selectivity. Despite an increasing understanding of the underlying mechanisms, application of this reaction remains limited by the cumulative performance trade-offs (i.e., click kinetics, release kinetics, release yield) of existing tools. Efficient release has been restricted to tetrazine scaffolds with comparatively low click reactivity, while highly reactive aryl-tetrazines give only minimal release. By introducing hydroxyl groups onto phenyl- and pyridyl-tetrazine scaffolds, we have developed a new class of 'bioorthogonal scissors' with unique chemical performance. We demonstrate that hydroxyaryl-tetrazines achieve near-quantitative release upon accelerated click reaction with cleavable trans-cyclooctenes, as exemplified by click-triggered activation of a caged prodrug, intramitochondrial cleavage of a fluorogenic probe (turn-on) in live cells, and rapid intracellular bioorthogonal disassembly (turn-off) of a ligand-dye conjugate.
Collapse
Affiliation(s)
- Martin Wilkovitsch
- Institute of Applied Synthetic Chemistry, TU Wien, 1060, Vienna, Austria
| | - Walter Kuba
- Institute of Applied Synthetic Chemistry, TU Wien, 1060, Vienna, Austria
| | - Patrick Keppel
- Institute of Applied Synthetic Chemistry, TU Wien, 1060, Vienna, Austria
| | - Barbara Sohr
- Institute of Applied Synthetic Chemistry, TU Wien, 1060, Vienna, Austria
| | - Andreas Löffler
- Institute of Applied Synthetic Chemistry, TU Wien, 1060, Vienna, Austria
| | - Stefan Kronister
- Institute of Applied Synthetic Chemistry, TU Wien, 1060, Vienna, Austria
| | - Andres Fernandez Del Castillo
- Institute of Applied Synthetic Chemistry, TU Wien, 1060, Vienna, Austria
- Center for Systems Biology & Department of Medicine, Massachusetts General Hospital, Harvard Medical School, 02114, Boston, MA, USA
| | - Marion Goldeck
- Institute of Applied Synthetic Chemistry, TU Wien, 1060, Vienna, Austria
- Center for Anatomy and Cell Biology, Medical University of Vienna, 1090, Vienna, Austria
| | - Rastislav Dzijak
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 16000, Prague 6, Czech Republic
| | - Michal Rahm
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 16000, Prague 6, Czech Republic
- University of Chemistry and Technology, Department of Chemistry of Natural Compounds, 16628, Prague 6, Czech Republic
| | - Milan Vrabel
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 16000, Prague 6, Czech Republic
| | - Dennis Svatunek
- Institute of Applied Synthetic Chemistry, TU Wien, 1060, Vienna, Austria
| | - Jonathan C T Carlson
- Center for Systems Biology & Department of Medicine, Massachusetts General Hospital, Harvard Medical School, 02114, Boston, MA, USA
| | - Hannes Mikula
- Institute of Applied Synthetic Chemistry, TU Wien, 1060, Vienna, Austria
| |
Collapse
|
12
|
Zeng T, Wu Q, Liu Y, Qi Q, Shen W, Gu W, Zhang Y, Xiong W, Xie Z, Qi X, Tian T, Zhou X. Unraveling the Cleavage Reaction of Hydroxylamines with Cyclopropenones Considering Biocompatibility. J Am Chem Soc 2024; 146:35077-35089. [PMID: 39660762 DOI: 10.1021/jacs.4c09757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
We develop a latent biocompatible cleavage reaction involving the hitherto unexplored interaction between hydroxylamines and cyclopropenones. Our study addresses the regioselectivity challenges commonly observed in asymmetric cyclopropenone transformations, substantiated by variations in substrate, Density Functional Theory calculations, and in situ NMR analysis. This reaction is characterized by high efficiency, broad substrate scope, stability, latent biocompatibility, and mild reaction conditions. Significantly, it facilitates fluorescence activation and functions as a controlled release mechanism for prodrugs, showing great promise in biological assays. Our success in achieving the controlled release of nitrogen mustard in HeLa cells underscores its potential application in cellular contexts. Additionally, we introduce a simple and highly efficient method for synthesizing α, β-substituted pentenolides, applicable to a variety of substrates. Moreover, we extend this cleavage reaction to the CRISPR-Cas9 system, achieving precise, on-demand regulation of guide RNA activity. The introduction of this cleavage reaction offers a promising tool for biochemical research and biotechnological applications.
Collapse
Affiliation(s)
- Tianying Zeng
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Quan Wu
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Yongjie Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Qianqian Qi
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Wei Shen
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Wei Gu
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Yuanyuan Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Wei Xiong
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Zhongpao Xie
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Xiaotian Qi
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Tian Tian
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| | - Xiang Zhou
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, The Institute of Molecular Medicine Medicine, Wuhan University People's Hospital, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, Hubei 430072, China
| |
Collapse
|
13
|
Versteegen RM, Rossin R, Filot IAW, Hoeben FJM, van Onzen AHAM, Janssen HM, Robillard MS. Ortho-functionalized pyridinyl-tetrazines break the inverse correlation between click reactivity and cleavage yields in click-to-release chemistry. Commun Chem 2024; 7:302. [PMID: 39702778 DOI: 10.1038/s42004-024-01392-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024] Open
Abstract
The bioorthogonal tetrazine-triggered cleavage of trans-cyclooctene(TCO)-linked payloads has strong potential for widespread use in drug delivery and in particular in click-cleavable antibody-drug conjugates (ADCs). However, clinical translation is hampered by an inverse correlation between click reactivity and payload release yield, requiring high doses of less reactive tetrazines to drive in vivo TCO reactions and payload release to completion. Herein we report that the cause for the low release when using the highly reactive bis-(2-pyridinyl)-tetrazine is the stability of the initially formed 4,5-dihydropyridazine product, precluding tautomerization to the releasing 1,4-dihydropyridazine tautomer. We demonstrate that efficient tautomerization and payload elimination can be achieved by ortho-substituting bis-pyridinyl-tetrazines with hydrogen-bonding hydroxyl or amido groups, achieving a.o. release yields of 96% with 18-fold more reactive tetrazines. Applied to on-tumor activation of a click-cleavable ADC in mice, these tetrazines afforded near-quantitative ADC conversion at a ca. 10- to 20-fold lower dose than what was previously needed, resulting in a strong therapeutic response.
Collapse
Affiliation(s)
- Ron M Versteegen
- SyMO-Chem B.V., Den Dolech 2, 5612 AZ, Eindhoven, The Netherlands
| | - Raffaella Rossin
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED, Nijmegen, The Netherlands
| | - Ivo A W Filot
- Eindhoven University of Technology, Den Dolech 2, 5612 AZ, Eindhoven, The Netherlands
| | - Freek J M Hoeben
- SyMO-Chem B.V., Den Dolech 2, 5612 AZ, Eindhoven, The Netherlands
| | | | - Henk M Janssen
- SyMO-Chem B.V., Den Dolech 2, 5612 AZ, Eindhoven, The Netherlands
| | - Marc S Robillard
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED, Nijmegen, The Netherlands.
| |
Collapse
|
14
|
Wang X, Wen S, Wu Z, Jiang JH. Orthogonal Control of Nucleic Acid Function via Chemical Caging-Decaging Strategies. Chembiochem 2024; 25:e202400516. [PMID: 39141545 DOI: 10.1002/cbic.202400516] [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: 06/15/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 08/16/2024]
Abstract
The ability to precisely control the function of nucleic acids plays an important role in biosensing and biomedicine. In recent years, novel strategies employing biological, physical, and chemical triggers have been developed to modulate the function of nucleic acids spatiotemporally. These approaches commonly involve the incorporation of stimuli-responsive groups onto nucleic acids to block their functions until triggers-induced decaging restore activity. These inventive strategies deepen our comprehension of nucleic acid molecules' dynamic behavior and provide new techniques for precise disease diagnosis and treatment. Focusing on the spatiotemporal regulation of nucleic acid molecules through the chemical caging-decaging strategy, we here present an overview of the innovative triggered control mechanisms and accentuate their implications across the fields of chemical biology, biomedicine, and biosensing.
Collapse
Affiliation(s)
- Xiangnan Wang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, School of Biomedical Science, Hunan University, Changsha, Hunan, 410082, P. R. China
- School of Resource & Environment, Hunan University of Technology and Business, Changsha, Hunan, 410082, P. R. China
| | - Siyu Wen
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, School of Biomedical Science, Hunan University, Changsha, Hunan, 410082, P. R. China
| | - Zhenkun Wu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, School of Biomedical Science, Hunan University, Changsha, Hunan, 410082, P. R. China
| | - Jian-Hui Jiang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, School of Biomedical Science, Hunan University, Changsha, Hunan, 410082, P. R. China
| |
Collapse
|
15
|
Charoenpattarapreeda J, Tegge W, Xu C, Harmrolfs K, Hinkelmann B, Wullenkord H, Hotop SK, Beutling U, Rox K, Brönstrup M. A Targeted Click-to-Release Activation of the Last-Resort Antibiotic Colistin Reduces its Renal Cell Toxicity. Angew Chem Int Ed Engl 2024; 63:e202408360. [PMID: 39113573 DOI: 10.1002/anie.202408360] [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: 05/02/2024] [Indexed: 10/17/2024]
Abstract
The use of highly potent but very toxic antibiotics such as colistin has become inevitable due to the rise of antimicrobial resistance. We aimed for a chemically-triggered, controlled release of colistin at the infection site to lower its systemic toxicity by harnessing the power of click-to-release reactions. Kinetic experiments with nine tetrazines and three dienophiles demonstrated a fast release via an inverse-electron-demand Diels-Alder reaction between trans-cyclooctene (TCO) and the amine-functionalised tetrazine Tz7. The antibiotic activity of colistin against Escherichia coli was masked by TCO units, but restored upon reaction with d-Ubi-Tz, a tetrazine functionalised with the bacterial binding peptide d-Ubi29-41. While standard TCO did not improve toxicity against human proximal tubular kidney HK-2 cells, the installation of an aspartic acid-modified TCO masking group reduced the overall charge of the peptide and entry to the kidney cells, thereby dramatically lowering its toxicity. The analog Col-(TCO-Asp)1 had favourable pharmacokinetic properties in mice and was successfully activated locally in the lung by d-Ubi-Tz in an in vivo infection model, whereas it remained inactive and non-harmful without the chemical trigger. This study constitutes the first example of a systemically acting two-component antibiotic with improved drug tolerability.
Collapse
Affiliation(s)
| | - Werner Tegge
- Department of Chemical Biology, Helmholtz-Zentrum für Infektionsforschung (HZI), Inhoffensstraße 7, 38124, Braunschweig, Germany
| | - Chunfa Xu
- Department of Chemical Biology, Helmholtz-Zentrum für Infektionsforschung (HZI), Inhoffensstraße 7, 38124, Braunschweig, Germany
| | - Kirsten Harmrolfs
- Department of Chemical Biology, Helmholtz-Zentrum für Infektionsforschung (HZI), Inhoffensstraße 7, 38124, Braunschweig, Germany
| | - Bettina Hinkelmann
- Department of Chemical Biology, Helmholtz-Zentrum für Infektionsforschung (HZI), Inhoffensstraße 7, 38124, Braunschweig, Germany
| | - Hannah Wullenkord
- Department of Chemical Biology, Helmholtz-Zentrum für Infektionsforschung (HZI), Inhoffensstraße 7, 38124, Braunschweig, Germany
| | - Sven-Kevin Hotop
- Department of Chemical Biology, Helmholtz-Zentrum für Infektionsforschung (HZI), Inhoffensstraße 7, 38124, Braunschweig, Germany
| | - Ulrike Beutling
- Department of Chemical Biology, Helmholtz-Zentrum für Infektionsforschung (HZI), Inhoffensstraße 7, 38124, Braunschweig, Germany
| | - Katharina Rox
- Department of Chemical Biology, Helmholtz-Zentrum für Infektionsforschung (HZI), Inhoffensstraße 7, 38124, Braunschweig, Germany
- Deutsche Zentrum für Infektionsforschung (DZIF), Site Hannover-Braunschweig, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz-Zentrum für Infektionsforschung (HZI), Inhoffensstraße 7, 38124, Braunschweig, Germany
- Deutsche Zentrum für Infektionsforschung (DZIF), Site Hannover-Braunschweig, Germany
- Biomolekulares Wirkstoffzentrum (BMWZ), Leibniz Universität Hannover, 30167, Hannover, Germany
| |
Collapse
|
16
|
Li J, Zhang T, Wu D, He C, Weng H, Zheng T, Liu J, Yao H, Chen J, Ren Y, Zhu Z, Xu J, Xu S. Palladium-Mediated Bioorthogonal System for Prodrug Activation of N-Benzylbenzamide-Containing Tubulin Polymerization Inhibitors for the Treatment of Solid Tumors. J Med Chem 2024; 67:19905-19924. [PMID: 39484713 DOI: 10.1021/acs.jmedchem.4c02419] [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: 11/03/2024]
Abstract
Bioorthogonal cleavage reactions have been developed as an intriguing strategy to enhance the safety of chemotherapeutics. Aiming to reduce the toxicity and improve the targeted release properties of the colchicine binding site inhibitors (CBSIs) based on previous work, a series of biologically inert prodrugs were further designed and synthesized through a bioorthogonal prodrug strategy. The therapeutic effects of prodrugs could be "turned-on" once combined with palladium resins. Particularly, prodrug 2b was 68.3-fold less cytotoxic compared to the parent compound, while its cytotoxicity was recovered in situ in the presence of palladium resins. Mechanism studies confirmed that 2b inhibited cell growth in the same manner as CBSIs. More importantly, in vivo efficacy studies demonstrated the efficient activation of 2b by palladium resins, resulting in significant inhibition of tumor growth (63.2%). These results suggest that prodrug 2b with improved safety and targeted release property catalyzed by a Pd-mediated bioorthogonal cleavage reaction deserves further investigation.
Collapse
Affiliation(s)
- Jinlong Li
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Tong Zhang
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Di Wu
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Chen He
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Haoxiang Weng
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Tiandong Zheng
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Jie Liu
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Hong Yao
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Jichao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Yansong Ren
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Zheying Zhu
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, The University of Nottingham, Nottingham NG7 2RD, U.K
| | - Jinyi Xu
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Shengtao Xu
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
- Department of Hepatobiliary Surgery, The First People's Hospital of Kunshan, Suzhou 215132, P.R. China
| |
Collapse
|
17
|
Bertolini M, Mendive-Tapia L, Karmakar U, Vendrell M. Chemo-Click: Receptor-Controlled and Bioorthogonal Chemokine Ligation for Real-Time Imaging of Drug-Resistant Leukemic B Cells. J Am Chem Soc 2024; 146:30565-30572. [PMID: 39441736 PMCID: PMC11544607 DOI: 10.1021/jacs.4c12035] [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: 08/30/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
Drug resistance in B cell leukemia is characterized by the coexpression of CXCR5 and CXCR3 chemokine receptors, making it a valuable biomarker for patient stratification. Herein, we report a novel platform of activatable chemokines to selectively image drug-resistant leukemic B cells for the first time. The C-terminal derivatization of the human chemokines CXCL13 and CXCL10 with bioorthogonal tetrazine-BODIPY and BCN groups retained binding and internalization via their cognate CXCR5 and CXCR3 receptors and enabled rapid fluorescence labeling of CXCR5+ CXCR3+ resistant B cells─but not drug-susceptible leukemic cells─via intracellular chemokine ligation. This modular chemical approach offers a versatile strategy for real-time immunophenotyping of cell populations with distinct chemokine profiles and will accelerate the design of new precision medicine tools to advance personalized therapies in blood tumors.
Collapse
Affiliation(s)
- Marco Bertolini
- Centre
for Inflammation Research, The University
of Edinburgh, Edinburgh EH16 4UU, U.K.
- IRR
Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, U.K.
| | - Lorena Mendive-Tapia
- Centre
for Inflammation Research, The University
of Edinburgh, Edinburgh EH16 4UU, U.K.
- IRR
Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, U.K.
| | - Utsa Karmakar
- Centre
for Inflammation Research, The University
of Edinburgh, Edinburgh EH16 4UU, U.K.
- IRR
Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, U.K.
| | - Marc Vendrell
- Centre
for Inflammation Research, The University
of Edinburgh, Edinburgh EH16 4UU, U.K.
- IRR
Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, U.K.
| |
Collapse
|
18
|
Liang T, Dong Y, Cheng I, Wen P, Li F, Liu F, Wu Q, Ren E, Liu P, Li H, Gu Z. In situ formation of biomolecular condensates as intracellular drug reservoirs for augmenting chemotherapy. Nat Biomed Eng 2024; 8:1469-1482. [PMID: 39271933 DOI: 10.1038/s41551-024-01254-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/10/2024] [Indexed: 09/15/2024]
Abstract
Biomolecular condensates, which arise from liquid-liquid phase separation within cells, may provide a means of enriching and prolonging the retention of small-molecule drugs within cells. Here we report a method for the controlled in situ formation of biomolecular condensates as reservoirs for the enrichment and retention of chemotherapeutics in cancer cells, and show that the approach can be leveraged to enhance antitumour efficacies in mice with drug-resistant tumours. The method involves histones as positively charged proteins and doxorubicin-intercalated DNA strands bioorthogonally linked via a click-to-release reaction between trans-cyclooctene and tetrazine groups. The reaction temporarily impaired the phase separation of histones in vitro, favoured the initiation of liquid-liquid phase separation within cells and led to the formation of biomolecular condensates that were sufficiently large to be retained within tumour cells. The controlled formation of biomolecular condensates as drug reservoirs within cells may offer new options for boosting the efficacies of cancer therapies.
Collapse
Affiliation(s)
- Tingxizi Liang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, and Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yuxiang Dong
- School of Materials Science and Engineering, Suzhou University of Science and Technology, Suzhou, China
| | - Irina Cheng
- State Key Laboratory of Advanced Drug Delivery and Release Systems, and Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ping Wen
- State Key Laboratory of Advanced Drug Delivery and Release Systems, and Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Fengqin Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Liu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, and Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qing Wu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, and Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - En Ren
- State Key Laboratory of Advanced Drug Delivery and Release Systems, and Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Peifeng Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Hongjun Li
- State Key Laboratory of Advanced Drug Delivery and Release Systems, and Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Zhen Gu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, and Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China.
| |
Collapse
|
19
|
Yao Y, Chen Y, Zhou C, Zhang Q, He X, Dong K, Yang C, Chu B, Qian Z. Bioorthogonal chemistry-based prodrug strategies for enhanced biosafety in tumor treatments: current progress and challenges. J Mater Chem B 2024; 12:10818-10834. [PMID: 39352785 DOI: 10.1039/d4tb01413h] [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: 10/04/2024]
Abstract
Cancer is a significant global health challenge, and while chemotherapy remains a widely used treatment, its non-specific toxicity and broad distribution can lead to systemic side effects and limit its effectiveness against tumors. Therefore, the development of safer chemotherapy alternatives is crucial. Prodrugs hold great promise, as they remain inactive until they reach the cancer site, where they are selectively activated by enzymes or specific factors, thereby reducing side effects and improving targeting. However, subtle differences in the microenvironments between tumors and normal tissue may still result in unintended cytotoxicity. Bioorthogonal reactions, known for their selectivity and precision without interfering with natural biochemical processes, are gaining attention. When combined with prodrug strategies, these reactions offer the potential to create highly effective chemotherapy drugs. This review examines the safety and efficacy of prodrug strategies utilizing various bioorthogonal reactions in cancer treatment.
Collapse
Affiliation(s)
- Yongchao Yao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
- Precision Medicine Translational Research Center (PMTRC), West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ying Chen
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China
| | - Chang Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Quanzhi Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Xun He
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Kai Dong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Chengli Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Bingyang Chu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Zhiyong Qian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
20
|
Yamada K, Mukaimine A, Nakamura A, Kusakari Y, Pradipta AR, Chang TC, Tanaka K. Chemistry-driven translocation of glycosylated proteins in mice. Nat Commun 2024; 15:7409. [PMID: 39358337 PMCID: PMC11446924 DOI: 10.1038/s41467-024-51342-5] [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: 11/02/2023] [Accepted: 08/06/2024] [Indexed: 10/04/2024] Open
Abstract
Cell surface glycans form various "glycan patterns" consisting of different types of glycan molecules, thus enabling strong and selective cell-to-cell recognition. We previously conjugated different N-glycans to human serum albumin to construct glycoalbumins mimicking natural glycan patterns that could selectively recognize target cells or control excretion pathways in mice. Here, we develop an innovative glycoalbumin capable of undergoing transformation and remodeling of its glycan pattern in vivo, which induces its translocation from the initial target to a second one. Replacing α(2,3)-sialylated N-glycans on glycoalbumin with galactosylated glycans induces the translocation of the glycoalbumin from blood or tumors to the intestine in mice. Such "in vivo glycan pattern remodeling" strategy can be used as a drug delivery system to promote excretion of a drug or medical radionuclide from the tumor after treatment, thereby preventing prolonged exposure leading to adverse effects. Alternatively, this study provides a potential strategy for using a single glycoalbumin for the simultaneous treatment of multiple diseases in a patient.
Collapse
Affiliation(s)
- Kenshiro Yamada
- 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
| | - Akari Mukaimine
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Akiko Nakamura
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Yuriko Kusakari
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Ambara R Pradipta
- 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
| | - Tsung-Che Chang
- 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 Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan.
| | - Katsunori Tanaka
- 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 Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan.
| |
Collapse
|
21
|
Niu W, Guo J. Cellular Site-Specific Incorporation of Noncanonical Amino Acids in Synthetic Biology. Chem Rev 2024; 124:10577-10617. [PMID: 39207844 PMCID: PMC11470805 DOI: 10.1021/acs.chemrev.3c00938] [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] [Indexed: 09/04/2024]
Abstract
Over the past two decades, genetic code expansion (GCE)-enabled methods for incorporating noncanonical amino acids (ncAAs) into proteins have significantly advanced the field of synthetic biology while also reaping substantial benefits from it. On one hand, they provide synthetic biologists with a powerful toolkit to enhance and diversify biological designs beyond natural constraints. Conversely, synthetic biology has not only propelled the development of ncAA incorporation through sophisticated tools and innovative strategies but also broadened its potential applications across various fields. This Review delves into the methodological advancements and primary applications of site-specific cellular incorporation of ncAAs in synthetic biology. The topics encompass expanding the genetic code through noncanonical codon addition, creating semiautonomous and autonomous organisms, designing regulatory elements, and manipulating and extending peptide natural product biosynthetic pathways. The Review concludes by examining the ongoing challenges and future prospects of GCE-enabled ncAA incorporation in synthetic biology and highlighting opportunities for further advancements in this rapidly evolving field.
Collapse
Affiliation(s)
- Wei Niu
- Department of Chemical & Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, United States
- The Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, United States
| | - Jiantao Guo
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, United States
- The Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, United States
| |
Collapse
|
22
|
Quintana J, Carlson JCT, Scott E, Ng TSC, Miller MA, Weissleder R. Scission-Enhanced Molecular Imaging (SEMI). Bioconjug Chem 2024; 35. [PMID: 39255972 PMCID: PMC11488501 DOI: 10.1021/acs.bioconjchem.4c00337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 09/12/2024]
Abstract
Positron emission tomography (PET) imaging methods have advanced our understanding of human biology, while targeted radiotherapeutic drug treatments are now routinely used clinically. The field is expected to grow considerably based on an expanding repertoire of available affinity ligands, radionuclides, conjugation chemistries, and their FDA approvals. With this increasing use, strategies for dose reduction have become of high interest to protect patients from unnecessary and off-target toxicity. Here, we describe a simple and powerful method, scission-enhanced molecular imaging (SEMI). The technique allows for rapid corporeal elimination of radionuclides once imaging or theranostic treatment is completed and relies on "click-to-release" bioorthogonal linkers.
Collapse
Affiliation(s)
- Jeremy
M. Quintana
- Center
for Systems Biology, Massachusetts General
Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Jonathan C. T. Carlson
- Center
for Systems Biology, Massachusetts General
Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
- Cancer
Center, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Ella Scott
- Center
for Systems Biology, Massachusetts General
Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
| | - Thomas S. C. Ng
- Center
for Systems Biology, Massachusetts General
Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Miles A. Miller
- Center
for Systems Biology, Massachusetts General
Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Ralph Weissleder
- Center
for Systems Biology, Massachusetts General
Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
- Cancer
Center, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Department
of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
23
|
Tomarchio EG, Turnaturi R, Saccullo E, Patamia V, Floresta G, Zagni C, Rescifina A. Tetrazine-trans-cyclooctene ligation: Unveiling the chemistry and applications within the human body. Bioorg Chem 2024; 150:107573. [PMID: 38905885 DOI: 10.1016/j.bioorg.2024.107573] [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: 02/16/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
Bioorthogonal reactions have revolutionized chemical biology by enabling selective chemical transformations within living organisms and cells. This review comprehensively explores bioorthogonal chemistry, emphasizing inverse-electron-demand Diels-Alder (IEDDA) reactions between tetrazines and strained dienophiles and their crucial role in chemical biology and various applications within the human body. This highly reactive and selective reaction finds diverse applications, including cleaving antibody-drug conjugates, prodrugs, proteins, peptide antigens, and enzyme substrates. The versatility extends to hydrogel chemistry, which is crucial for biomedical applications, yet it faces challenges in achieving precise cellularization. In situ activation of cytotoxic compounds from injectable biopolymer belongs to the click-activated protodrugs against cancer (CAPAC) platform, an innovative approach to tumor-targeted prodrug delivery and activation. The CAPAC platform, relying on click chemistry between trans-cyclooctene (TCO) and tetrazine-modified biopolymers, exhibits modularity across diverse tumor characteristics, presenting a promising approach in anticancer therapeutics. The review highlights the importance of bioorthogonal reactions in developing radiopharmaceuticals for positron emission tomography (PET) imaging and theranostics, offering a promising avenue for diverse therapeutic applications.
Collapse
Affiliation(s)
- Elisabetta Grazia Tomarchio
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy; Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| | - Rita Turnaturi
- Institute of Cristallography CNR-IC, Via Paolo Gaifami 18, 95126 Catania, Italy
| | - Erika Saccullo
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy; Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| | - Vincenzo Patamia
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy
| | - Giuseppe Floresta
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy
| | - Chiara Zagni
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy.
| | - Antonio Rescifina
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
24
|
Yang H, Sun H, Chen Y, Wang Y, Yang C, Yuan F, Wu X, Chen W, Yin P, Liang Y, Wu H. Enabling Universal Access to Rapid and Stable Tetrazine Bioorthogonal Probes through Triazolyl-Tetrazine Formation. JACS AU 2024; 4:2853-2861. [PMID: 39211625 PMCID: PMC11350731 DOI: 10.1021/jacsau.3c00843] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/07/2024] [Accepted: 05/16/2024] [Indexed: 09/04/2024]
Abstract
Despite the immense potential of tetrazine bioorthogonal chemistry in biomedical research, the in vivo performance of tetrazine probes is challenged by the inverse correlation between the physiological stability and reactivity of tetrazines. Additionally, the synthesis of functionalized tetrazines is often complex and requires specialized reagents. To overcome these issues, we present a novel tetrazine scaffold-triazolyl-tetrazine-that can be readily synthesized from shelf-stable ethynyl-tetrazines and azides. Triazolyl-tetrazines exhibit improved physiological stability along with high reactivity. We showcase the effectiveness of this approach by creating cell-permeable probes for protein labeling and live cell imaging, as well as efficiently producing 18F-labeled molecular probes for positron emission tomography imaging. By utilizing the readily available pool of functionalized azides, we envisage that this modular approach will provide universal accessibility to tetrazine bioorthogonal tools, facilitating applications in biomedicine and materials science.
Collapse
Affiliation(s)
- Haojie Yang
- Department
of Radiology and Huaxi MR Research Center, Functional and Molecular
Imaging Key Laboratory of Sichuan Province and Frontiers Science Center
for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongbao Sun
- Department
of Radiology and Huaxi MR Research Center, Functional and Molecular
Imaging Key Laboratory of Sichuan Province and Frontiers Science Center
for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yinghan Chen
- State
Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of
Advanced Organic Materials, School of Chemistry and Chemical Engineering,
Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| | - Yayue Wang
- Department
of Radiology and Huaxi MR Research Center, Functional and Molecular
Imaging Key Laboratory of Sichuan Province and Frontiers Science Center
for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Cheng Yang
- Key
Laboratory of Drug-Targeting and Drug Delivery System of the Education
Ministry and Sichuan Province, Sichuan University, Chengdu 610041, China
| | - Fang Yuan
- Department
of Radiology and Huaxi MR Research Center, Functional and Molecular
Imaging Key Laboratory of Sichuan Province and Frontiers Science Center
for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoai Wu
- Department
of Nuclear Medicine and Clinical Nuclear Medicine Research Lab, West
China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Chen
- Department
of Nuclear Medicine and Clinical Nuclear Medicine Research Lab, West
China Hospital, Sichuan University, Chengdu 610041, China
| | - Ping Yin
- School
of Materials Science & Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Yong Liang
- State
Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of
Advanced Organic Materials, School of Chemistry and Chemical Engineering,
Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| | - Haoxing Wu
- Department
of Radiology and Huaxi MR Research Center, Functional and Molecular
Imaging Key Laboratory of Sichuan Province and Frontiers Science Center
for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
- Key
Laboratory of Drug-Targeting and Drug Delivery System of the Education
Ministry and Sichuan Province, Sichuan University, Chengdu 610041, China
| |
Collapse
|
25
|
Pandit B, Fang L, Kool ET, Royzen M. Reversible RNA Acylation Using Bio-Orthogonal Chemistry Enables Temporal Control of CRISPR-Cas9 Nuclease Activity. ACS Chem Biol 2024; 19:1719-1724. [PMID: 39051564 PMCID: PMC11334111 DOI: 10.1021/acschembio.4c00117] [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: 02/16/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
The CRISPR-Cas9 system is a widely popular tool for genome engineering. There is strong interest in developing tools for temporal control of CRISPR-Cas9 activity to address some of the challenges and to broaden the scope of potential applications. In this work, we describe a bio-orthogonal chemistry-based approach to control nuclease activity with temporal precision. We report a trans-cyclooctene (TCO)-acylimidazole reagent that acylates 2'-OH groups of RNA. Poly acylation ("cloaking") of RNA was optimized in vitro using a model 18-nt oligonucleotide, as well as CRISPR single guide RNA (sgRNA). Two hours of treatment completely inactivated sgRNA for Cas9-assisted DNA cleavage. Nuclease activity was restored upon addition of tetrazine, which removes the TCO moieties via a two-step process ("uncloaking"). The approach was applied to target the GFP gene in live HEK293 cells. GFP expression was analyzed by flow cytometry. In the future, we anticipate that our approach will be useful in the field of developmental biology, by enabling investigation of genes of interest at different stages of an organism's development.
Collapse
Affiliation(s)
- Bhoomika Pandit
- Department
of Chemistry, University at Albany, 1400 Washington Ave, Albany, New York 12222, United States
| | - Linglan Fang
- Department
of Chemistry, Stanford University, 450 Serra Mall, Stanford, California 94305, United States
| | - Eric T. Kool
- Department
of Chemistry, Stanford University, 450 Serra Mall, Stanford, California 94305, United States
| | - Maksim Royzen
- Department
of Chemistry, University at Albany, 1400 Washington Ave, Albany, New York 12222, United States
| |
Collapse
|
26
|
Knaack JIH, Meier C. Out of the Dark, into the Light: Metabolic Fluorescent Labeling of Nucleic Acids. ChemMedChem 2024; 19:e202400160. [PMID: 38712684 DOI: 10.1002/cmdc.202400160] [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: 02/29/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/08/2024]
Abstract
This review outlines recent advances in live-cell imaging techniques for nucleic acids. We describe the evolution of these methods, particularly highlighting the development of metabolic labeling approaches compatible with living systems using fluorescence-based labeling.
Collapse
Affiliation(s)
- J Iven H Knaack
- Department of Chemistry, Faculty of Sciences, University of Hamburg, Martin-Luther-King-Platz 6, 20146, Hamburg, Germany
| | - Chris Meier
- Department of Chemistry, Faculty of Sciences, University of Hamburg, Martin-Luther-King-Platz 6, 20146, Hamburg, Germany
| |
Collapse
|
27
|
Zhao R, Chen Y, Liang Y. Bioorthogonal Delivery of Carbon Disulfide in Living Cells. Angew Chem Int Ed Engl 2024; 63:e202400020. [PMID: 38752888 DOI: 10.1002/anie.202400020] [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: 01/01/2024] [Indexed: 06/27/2024]
Abstract
Carbon disulfide (CS2) is an environmental contaminant, which is deadly hazardous to the workers under chronic or acute exposure. However, the toxicity mechanisms of CS2 are still unclear due to the scarcity of biocompatible donors, which can release CS2 in cells. Here we developed the first bioorthogonal CS2 delivery system based on the "click-and-release" reactions between mesoionic 1,3-thiazolium-5-thiolates (TATs) and strained cyclooctyne exo-BCN-OH. We successfully realized intracellular CS2 release and investigated the causes of CS2-induced hepatotoxicity, including oxidative stress, proteotoxic stress and copper-dependent cell death. It is found that CS2 can be copper vehicles bypassing copper transporters after reacting with nucleophiles in cytoplasm, and extra copper supplementation will exacerbate the loss of homeostasis of cells and ultimately cell death. These findings inspired us to explore the anticancer activity of CS2 in combination with copper by introducing a copper chelating group in our CS2 delivery system.
Collapse
Affiliation(s)
- Ruohan Zhao
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yinghan Chen
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yong Liang
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
28
|
Adhikari P, Li G, Go M, Mandikian D, Rafidi H, Ng C, Anifa S, Johnson K, Bao L, Hernandez Barry H, Rowntree R, Agard N, Wu C, Chou KJ, Zhang D, Kozak KR, Pillow TH, Lewis GD, Yu SF, Boswell CA, Sadowsky JD. On Demand Bioorthogonal Switching of an Antibody-Conjugated SPECT Probe to a Cytotoxic Payload: from Imaging to Therapy. J Am Chem Soc 2024; 146:19088-19100. [PMID: 38946086 DOI: 10.1021/jacs.4c03529] [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: 07/02/2024]
Abstract
Antibody-drug conjugates (ADCs) for the treatment of cancer aim to achieve selective delivery of a cytotoxic payload to tumor cells while sparing normal tissue. In vivo, multiple tumor-dependent and -independent processes act on ADCs and their released payloads to impact tumor-versus-normal delivery, often resulting in a poor therapeutic window. An ADC with a labeled payload would make synchronous correlations between distribution and tissue-specific pharmacological effects possible, empowering preclinical and clinical efforts to improve tumor-selective delivery; however, few methods to label small molecules without destroying their pharmacological activity exist. Herein, we present a bioorthogonal switch approach that allows a radiolabel attached to an ADC payload to be removed tracelessly at will. We exemplify this approach with a potent DNA-damaging agent, the pyrrolobenzodiazepine (PBD) dimer, delivered as an antibody conjugate targeted to lung tumor cells. The radiometal chelating group, DOTA, was attached via a novel trans-cyclooctene (TCO)-caged self-immolative para-aminobenzyl (PAB) linker to the PBD, stably attenuating payload activity and allowing tracking of biodistribution in tumor-bearing mice via SPECT-CT imaging (live) or gamma counting (post-mortem). Following TCO-PAB-DOTA reaction with tetrazines optimized for extra- and intracellular reactivity, the label was removed to reveal the unmodified PBD dimer capable of inducing potent tumor cell killing in vitro and in mouse xenografts. The switchable antibody radio-drug conjugate (ArDC) we describe integrates, but decouples, the two functions of a theranostic given that it can serve as a diagnostic for payload delivery in the labeled state, but can be switched on demand to a therapeutic agent (an ADC).
Collapse
Affiliation(s)
- Pragya Adhikari
- Genentech Inc., South San Francisco, California 94080, United States
| | - Guangmin Li
- Genentech Inc., South San Francisco, California 94080, United States
| | - MaryAnn Go
- Genentech Inc., South San Francisco, California 94080, United States
| | | | - Hanine Rafidi
- Genentech Inc., South San Francisco, California 94080, United States
| | - Carl Ng
- Genentech Inc., South San Francisco, California 94080, United States
| | - Sagana Anifa
- Genentech Inc., South San Francisco, California 94080, United States
| | - Kevin Johnson
- Genentech Inc., South San Francisco, California 94080, United States
| | - Linda Bao
- Genentech Inc., South San Francisco, California 94080, United States
| | | | - Rebecca Rowntree
- Genentech Inc., South San Francisco, California 94080, United States
| | - Nicholas Agard
- Genentech Inc., South San Francisco, California 94080, United States
| | - Cong Wu
- Genentech Inc., South San Francisco, California 94080, United States
| | - Kang-Jye Chou
- Genentech Inc., South San Francisco, California 94080, United States
| | - Donglu Zhang
- Genentech Inc., South San Francisco, California 94080, United States
| | - Katherine R Kozak
- Genentech Inc., South San Francisco, California 94080, United States
| | - Thomas H Pillow
- Genentech Inc., South San Francisco, California 94080, United States
| | - Gail D Lewis
- Genentech Inc., South San Francisco, California 94080, United States
| | - Shang-Fan Yu
- Genentech Inc., South San Francisco, California 94080, United States
| | - C Andrew Boswell
- Genentech Inc., South San Francisco, California 94080, United States
| | - Jack D Sadowsky
- Genentech Inc., South San Francisco, California 94080, United States
| |
Collapse
|
29
|
Wu C, Xie J, Yao Q, Song Y, Yang G, Zhao J, Zhang R, Wang T, Jiang X, Cai X, Gao Y. Intrahippocampal Supramolecular Assemblies Directed Bioorthogonal Liberation of Neurotransmitters to Suppress Seizures in Freely Moving Mice. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2314310. [PMID: 38655719 DOI: 10.1002/adma.202314310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/22/2024] [Indexed: 04/26/2024]
Abstract
The precise delivery of anti-seizure medications (ASM) to epileptic loci remains the major challenge to treat epilepsy without causing adverse drug reactions. The unprovoked nature of epileptic seizures raises the additional need to release ASMs in a spatiotemporal controlled manner. Targeting the oxidative stress in epileptic lesions, here the reactive oxygen species (ROS) induced in situ supramolecular assemblies that synergized bioorthogonal reactions to deliver inhibitory neurotransmitter (GABA) on-demand, are developed. Tetrazine-bearing assembly precursors undergo oxidation and selectively self-assemble under pathological conditions inside primary neurons and mice brains. Assemblies induce local accumulation of tetrazine in the hippocampus CA3 region, which allows the subsequent bioorthogonal release of inhibitory neurotransmitters. For induced acute seizures, the sustained release of GABA extends the suppression than the direct supply of GABA. In the model of permanent damage of CA3, bioorthogonal ligation on assemblies provides a reservoir of GABA that behaves prompt release upon 365 nm irradiation. Incorporated with the state-of-the-art microelectrode arrays, it is elucidated that the bioorthogonal release of GABA shifts the neuron spike waveforms to suppress seizures at the single-neuron precision. The strategy of in situ supramolecular assemblies-directed bioorthogonal prodrug activation shall be promising for the effective delivery of ASMs to treat epilepsy.
Collapse
Affiliation(s)
- Chengling Wu
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
- CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Jingyu Xie
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qingxin Yao
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
- CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Yilin Song
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Gucheng Yang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jie Zhao
- CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Ruijia Zhang
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
- CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Ting Wang
- CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Xingyu Jiang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Xinxia Cai
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuan Gao
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
- CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, China
| |
Collapse
|
30
|
Luu T, Gristwood K, Knight JC, Jörg M. Click Chemistry: Reaction Rates and Their Suitability for Biomedical Applications. Bioconjug Chem 2024; 35:715-731. [PMID: 38775705 PMCID: PMC11191409 DOI: 10.1021/acs.bioconjchem.4c00084] [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: 02/25/2024] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 06/21/2024]
Abstract
Click chemistry has become a commonly used synthetic method due to the simplicity, efficiency, and high selectivity of this class of chemical reactions. Since their initial discovery, further click chemistry methods have been identified and added to the toolbox of click chemistry reactions for biomedical applications. However, selecting the most suitable reaction for a specific application is often challenging, as multiple factors must be considered, including selectivity, reactivity, biocompatibility, and stability. Thus, this review provides an overview of the benefits and limitations of well-established click chemistry reactions with a particular focus on the importance of considering reaction rates, an often overlooked criterion with little available guidance. The importance of understanding each click chemistry reaction beyond simply the reaction speed is discussed comprehensively with reference to recent biomedical research which utilized click chemistry. This review aims to provide a practical resource for researchers to guide the selection of click chemistry classes for different biomedical applications.
Collapse
Affiliation(s)
- Tracey Luu
- Medicinal
Chemistry Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Katie Gristwood
- School
of Natural & Environmental Sciences, Newcastle University, Newcastle Upon Tyne NE1 7RU, U.K.
| | - James C. Knight
- School
of Natural & Environmental Sciences, Newcastle University, Newcastle Upon Tyne NE1 7RU, U.K.
| | - Manuela Jörg
- Medicinal
Chemistry Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- School
of Natural & Environmental Sciences, Newcastle University, Newcastle Upon Tyne NE1 7RU, U.K.
| |
Collapse
|
31
|
Yi HB, Lee S, Seo K, Kim H, Kim M, Lee HS. Cellular and Biophysical Applications of Genetic Code Expansion. Chem Rev 2024; 124:7465-7530. [PMID: 38753805 DOI: 10.1021/acs.chemrev.4c00112] [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: 05/18/2024]
Abstract
Despite their diverse functions, proteins are inherently constructed from a limited set of building blocks. These compositional constraints pose significant challenges to protein research and its practical applications. Strategically manipulating the cellular protein synthesis system to incorporate novel building blocks has emerged as a critical approach for overcoming these constraints in protein research and application. In the past two decades, the field of genetic code expansion (GCE) has achieved significant advancements, enabling the integration of numerous novel functionalities into proteins across a variety of organisms. This technological evolution has paved the way for the extensive application of genetic code expansion across multiple domains, including protein imaging, the introduction of probes for protein research, analysis of protein-protein interactions, spatiotemporal control of protein function, exploration of proteome changes induced by external stimuli, and the synthesis of proteins endowed with novel functions. In this comprehensive Review, we aim to provide an overview of cellular and biophysical applications that have employed GCE technology over the past two decades.
Collapse
Affiliation(s)
- Han Bin Yi
- Department of Chemistry, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| | - Seungeun Lee
- Department of Chemistry, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| | - Kyungdeok Seo
- Department of Chemistry, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| | - Hyeongjo Kim
- Department of Chemistry, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| | - Minah Kim
- Department of Chemistry, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| | - Hyun Soo Lee
- Department of Chemistry, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| |
Collapse
|
32
|
Adhikari K, Vanermen M, Da Silva G, Van den Wyngaert T, Augustyns K, Elvas F. Trans-cyclooctene-a Swiss army knife for bioorthogonal chemistry: exploring the synthesis, reactivity, and applications in biomedical breakthroughs. EJNMMI Radiopharm Chem 2024; 9:47. [PMID: 38844698 PMCID: PMC11156836 DOI: 10.1186/s41181-024-00275-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/27/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Trans-cyclooctenes (TCOs) are highly strained alkenes with remarkable reactivity towards tetrazines (Tzs) in inverse electron-demand Diels-Alder reactions. Since their discovery as bioorthogonal reaction partners, novel TCO derivatives have been developed to improve their reactivity, stability, and hydrophilicity, thus expanding their utility in diverse applications. MAIN BODY TCOs have garnered significant interest for their applications in biomedical settings. In chemical biology, TCOs serve as tools for bioconjugation, enabling the precise labeling and manipulation of biomolecules. Moreover, their role in nuclear medicine is substantial, with TCOs employed in the radiolabeling of peptides and other biomolecules. This has led to their utilization in pretargeted nuclear imaging and therapy, where they function as both bioorthogonal tags and radiotracers, facilitating targeted disease diagnosis and treatment. Beyond these applications, TCOs have been used in targeted cancer therapy through a "click-to-release" approach, in which they act as key components to selectively deliver therapeutic agents to cancer cells, thereby enhancing treatment efficacy while minimizing off-target effects. However, the search for a suitable TCO scaffold with an appropriate balance between stability and reactivity remains a challenge. CONCLUSIONS This review paper provides a comprehensive overview of the current state of knowledge regarding the synthesis of TCOs, and its challenges, and their development throughout the years. We describe their wide ranging applications as radiolabeled prosthetic groups for radiolabeling, as bioorthogonal tags for pretargeted imaging and therapy, and targeted drug delivery, with the aim of showcasing the versatility and potential of TCOs as valuable tools in advancing biomedical research and applications.
Collapse
Affiliation(s)
- Karuna Adhikari
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
- Molecular Imaging and Radiology, University of Antwerp, Antwerp, Belgium
| | - Maarten Vanermen
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
- Molecular Imaging and Radiology, University of Antwerp, Antwerp, Belgium
| | - Gustavo Da Silva
- Molecular Imaging and Radiology, University of Antwerp, Antwerp, Belgium
| | - Tim Van den Wyngaert
- Molecular Imaging and Radiology, University of Antwerp, Antwerp, Belgium
- Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Koen Augustyns
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium.
| | - Filipe Elvas
- Molecular Imaging and Radiology, University of Antwerp, Antwerp, Belgium.
- Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium.
| |
Collapse
|
33
|
Mitry MMA, Dallas ML, Boateng SY, Greco F, Osborn HMI. Selective activation of prodrugs in breast cancer using metabolic glycoengineering and the tetrazine ligation bioorthogonal reaction. Bioorg Chem 2024; 147:107304. [PMID: 38643563 DOI: 10.1016/j.bioorg.2024.107304] [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: 01/25/2024] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 04/23/2024]
Abstract
Increasing the selectivity of chemotherapies by converting them into prodrugs that can be activated at the tumour site decreases their side effects and allows discrimination between cancerous and non-cancerous cells. Herein, the use of metabolic glycoengineering (MGE) to selectively label MCF-7 breast cancer cells with tetrazine (Tz) activators for subsequent activation of prodrugs containing the trans-cyclooctene (TCO) moiety by a bioorthogonal reaction is demonstrated. Three novel Tz-modified monosaccharides, Ac4ManNTz 7, Ac4GalNTz 8, and Ac4SiaTz 16, were used for expression of the Tz activator within sialic-acid rich breast cancer cells' surface glycans through MGE. Tz expression on breast cancer cells (MCF-7) was evaluated versus the non-cancerous L929 fibroblasts showing a concentration-dependant effect and excellent selectivity with ≥35-fold Tz expression on the MCF-7 cells versus the non-cancerous L929 fibroblasts. Next, a novel TCO-N-mustard prodrug and a TCO-doxorubicin prodrug were analyzed in vitro on the Tz-bioengineered cells to probe our hypothesis that these could be activated via a bioorthogonal reaction. Selective prodrug activation and restoration of cytotoxicity were demonstrated for the MCF-7 breast cancer cells versus the non-cancerous L929 cells. Restoration of the parent drug's cytotoxicity was shown to be dependent on the level of Tz expression where the Ac4ManNTz 7 and Ac4GalNTz 8 derivatives (20 µM) lead to the highest Tz expression and full restoration of the parent drug's cytotoxicity. This work suggests the feasibility of combining MGE and tetrazine ligation for selective prodrug activation in breast cancer.
Collapse
Affiliation(s)
- Madonna M A Mitry
- Reading School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AD. UK; Dept. of Pharmaceutical Chemistry, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt.
| | - Mark L Dallas
- Reading School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AD. UK.
| | - Samuel Y Boateng
- School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6UB, UK.
| | - Francesca Greco
- Reading School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AD. UK.
| | - Helen M I Osborn
- Reading School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AD. UK.
| |
Collapse
|
34
|
Xu C, Xu M, Hu Y, Liu J, Cheng P, Zeng Z, Pu K. Ingestible Artificial Urinary Biomarker Probes for Urine Test of Gastrointestinal Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2314084. [PMID: 38446383 DOI: 10.1002/adma.202314084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 03/05/2024] [Indexed: 03/07/2024]
Abstract
Although colorectal cancer diagnosed at an early stage shows high curability, methods simultaneously possessing point-of-care testing ability and high sensitivity are limited. Here, an orally deliverable biomarker-activatable probe (termed as HATS) for early detection of orthotopic tumors via remote urinalysis is presented. To enable its oral delivery to the colon, HATS is designed to have remarkable resistance to acidity and digestive enzymes in the stomach and small intestine and negligible intestinal absorption. Upon reaction with a cancer biomarker in the colon segment, HATS releases a small fragment of tetrazine that can transverse the intestinal barrier, enter blood circulation, and ultimately undergo renal clearance to urine. Subsequently, the urinary tetrazine fragment is detected by bioorthogonal reaction with trans-cyclooctene-caged resorufin (TCO-Reso) to afford a rapid and specific fluorescence enhancement of TCO-Reso. Such signal readout is correlated with the urinary tetrazine concentration and thus measures the level of cancer biomarkers in the colon. HATS-based optical urinalysis detects orthotopic colon tumors two weeks earlier than clinical serological tests and can be developed to a point-of-care paper test. Thereby, HATS-based urinalysis provides a non-invasive and sensitive approach to cancer screening at low-resource settings.
Collapse
Affiliation(s)
- Cheng Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Mengke Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Yuxuan Hu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Jing Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Penghui Cheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Ziling Zeng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921, Singapore
| |
Collapse
|
35
|
He X, Li J, Liang X, Mao W, Deng X, Qin M, Su H, Wu H. An all-in-one tetrazine reagent for cysteine-selective labeling and bioorthogonal activable prodrug construction. Nat Commun 2024; 15:2831. [PMID: 38565562 PMCID: PMC10987521 DOI: 10.1038/s41467-024-47188-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 03/25/2024] [Indexed: 04/04/2024] Open
Abstract
The prodrug design strategy offers a potent solution for improving therapeutic index and expanding drug targets. However, current prodrug activation designs are mainly responsive to endogenous stimuli, resulting in unintended drug release and systemic toxicity. In this study, we introduce 3-vinyl-6-oxymethyl-tetrazine (voTz) as an all-in-one reagent for modular preparation of tetrazine-caged prodrugs and chemoselective labeling peptides to produce bioorthogonal activable peptide-prodrug conjugates. These stable prodrugs can selectively bind to target cells, facilitating cellular uptake. Subsequent bioorthogonal cleavage reactions trigger prodrug activation, significantly boosting potency against tumor cells while maintaining exceptional off-target safety for normal cells. In vivo studies demonstrate the therapeutic efficacy and safety of this prodrug design approach. Given the broad applicability of functional groups and labeling versatility with voTz, we foresee that this strategy will offer a versatile solution to enhance the therapeutic range of cytotoxic agents and facilitate the development of bioorthogonal activatable biopharmaceuticals and biomaterials.
Collapse
Affiliation(s)
- Xinyu He
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Li
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xinxin Liang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Wuyu Mao
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xinglong Deng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan University, Chengdu, China
| | - Meng Qin
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Su
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Haoxing Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan University, Chengdu, China.
| |
Collapse
|
36
|
Zhu Y, Ding W, Chen Y, Shan Y, Liu C, Fan X, Lin S, Chen PR. Genetically encoded bioorthogonal tryptophan decaging in living cells. Nat Chem 2024; 16:533-542. [PMID: 38418535 DOI: 10.1038/s41557-024-01463-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 01/29/2024] [Indexed: 03/01/2024]
Abstract
Tryptophan (Trp) plays a critical role in the regulation of protein structure, interactions and functions through its π system and indole N-H group. A generalizable method for blocking and rescuing Trp interactions would enable the gain-of-function manipulation of various Trp-containing proteins in vivo, but generating such a platform remains challenging. Here we develop a genetically encoded N1-vinyl-caged Trp capable of rapid and bioorthogonal decaging through an optimized inverse electron-demand Diels-Alder reaction, allowing site-specific activation of Trp on a protein of interest in living cells. This chemical activation of a genetically encoded caged-tryptophan (Trp-CAGE) strategy enables precise activation of the Trp of interest underlying diverse important molecular interactions. We demonstrate the utility of Trp-CAGE across various protein families, such as catalase-peroxidases and kinases, as translation initiators and posttranslational modification readers, allowing the modulation of epigenetic signalling in a temporally controlled manner. Coupled with computer-aided prediction, our strategy paves the way for bioorthogonal Trp activation on more than 28,000 candidate proteins within their native cellular settings.
Collapse
Affiliation(s)
- Yuchao Zhu
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Wenlong Ding
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yulin Chen
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, China
| | - Ye Shan
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Chao Liu
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, China
| | - Xinyuan Fan
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China.
| | - Shixian Lin
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, China.
- Department of Medical Oncology, State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Peng R Chen
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
37
|
Yan X, Li K, Xie TQ, Jin XK, Zhang C, Li QR, Feng J, Liu CJ, Zhang XZ. Bioorthogonal "Click and Release" Reaction-Triggered Aggregation of Gold Nanoparticles Combined with Released Lonidamine for Enhanced Cancer Photothermal Therapy. Angew Chem Int Ed Engl 2024; 63:e202318539. [PMID: 38303647 DOI: 10.1002/anie.202318539] [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: 12/03/2023] [Revised: 01/26/2024] [Accepted: 02/01/2024] [Indexed: 02/03/2024]
Abstract
Cancer has been the most deadly disease, and 13 million cancer casualties are estimated to occur each year by 2030. Gold nanoparticles (AuNPs)-based photothermal therapy (PTT) has attracted great interest due to its high spatiotemporal controllability and noninvasiveness. Due to the trade-off between particle size and photothermal efficiency of AuNPs, rational design is needed to realize aggregation of AuNPs into larger particles with desirable NIR adsorption in tumor site. Exploiting the bioorthogonal "Click and Release" (BCR) reaction between iminosydnone and cycloalkyne, aggregation of AuNPs can be achieved and attractively accompanied by the release of chemotherapeutic drug purposed to photothermal synergizing. We synthesize iminosydnone-lonidamine (ImLND) as a prodrug and choose dibenzocyclooctyne (DBCO) as the trigger of BCR reaction. A PEGylated AuNPs-based two-component nanoplatform consisting of prodrug-loaded AuNPs-ImLND and tumor-targeting peptide RGD-conjugated AuNPs-DBCO-RGD is designed. In the therapeutic regimen, AuNPs-DBCO-RGD are intravenously injected first for tumor-specific enrichment and retention. Once the arrival of AuNPs-ImLND injected later at tumor site, highly photothermally active nanoaggregates of AuNPs are formed via the BCR reaction between ImLND and DBCO. The simultaneous release of lonidamine further enhanced the therapeutic performance by sensitizing cancer cells to PTT.
Collapse
Affiliation(s)
- Xiao Yan
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Ke Li
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Tian-Qiu Xie
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xiao-Kang Jin
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Cheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Qian-Ru Li
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Jun Feng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Chuan-Jun Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| |
Collapse
|
38
|
Yang C, Tripathi R, Wang B. Click chemistry in the development of PROTACs. RSC Chem Biol 2024; 5:189-197. [PMID: 38456041 PMCID: PMC10915971 DOI: 10.1039/d3cb00199g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 12/16/2023] [Indexed: 03/09/2024] Open
Abstract
Proteolysis-targeting chimeras or PROTACs are hetero-bifunctional molecules designed to mediate the disposal of a target protein via recruitment of the ubiquitination-proteasome degradation machinery. Because of the chimeric nature of such molecules, their synthesis requires a key step of "assembling" whether in the lab or in situ. Furthermore, targeted PROTACs often are hetero-trifunctional and require a second "assembling" step. Click chemistry has the unique advantages of tethering two or more molecular entities of choice under near physiological conditions and therefore has been applied to the development of PROTACs in various ways. This review provides a succinct summary of this field with a critical analysis of various factors that need to be considered for optimal results. Specifically, we examine issues including applications of click chemistry in in situ assembly for improved delivery, conjugation with a targeting group for selectivity, rapid synthesis for linker optimization, and lysosomal degradation of extracellular and membrane-associated proteins. We also examine reaction kinetics issues whenever possible or warranted.
Collapse
Affiliation(s)
- Ce Yang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University Atlanta Georgia 30303 USA
| | - Ravi Tripathi
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University Atlanta Georgia 30303 USA
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University Atlanta Georgia 30303 USA
| |
Collapse
|
39
|
Chang M, Dong Y, Xu H, Cruickshank-Taylor AB, Kozora JS, Behpour B, Wang W. Senolysis Enabled by Senescent Cell-Sensitive Bioorthogonal Tetrazine Ligation. Angew Chem Int Ed Engl 2024; 63:e202315425. [PMID: 38233359 PMCID: PMC11226389 DOI: 10.1002/anie.202315425] [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: 10/12/2023] [Revised: 01/11/2024] [Accepted: 01/17/2024] [Indexed: 01/19/2024]
Abstract
Although the clearance of senescent cells has been proven to slow down the aging process and promote anti-cancer chemotherapy, the development of senolytics remains challenging. Herein, we report a senolytic strategy enabled by senescent cell-sensitive bioorthogonal tetrazine ligation. Our design is based on linking dihydrotetrazine (Tz) to a galactose (Gal) moiety that serves both as a recognition moiety for senescence-associated β-galactosidase and a caging group for the control of tetrazine activity. Gal-Tz enables efficient click-release of a fluorescent hemicyanine and doxorubicin from a trans-cyclooctene-caged prodrug to detect and eliminate senescent HeLa and A549 cells over non-senescent counterparts with a 16.44 senolytic index. Furthermore, we leverage the strategy for the selective activation and delivery of proteolysis-targeting chimeras (PROTACs) as senolytics. PROTAC prodrug TCO-ARV-771 can be selectively activated by Gal-Tz and delivered into senescent HeLa and A549 cells to induce the degradation of bromodomain-containing protein 4. Senolytic PROTACs may offer an efficient way for intervention on cell senescence thanks to their unique capacity to degrade target proteins in a sub-stoichiometric and catalytic fashion. The results of this study establish the bioorthogonal tetrazine ligation approach as a viable strategy for selective removal of senescent cells.
Collapse
Affiliation(s)
- Mengyang Chang
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, 85721, USA
| | - Yue Dong
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona, 85721, USA
| | - Hang Xu
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona, 85721, USA
| | | | - Jacob S Kozora
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona, 85721, USA
| | - Baran Behpour
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, 85721, USA
| | - Wei Wang
- Departments of Pharmacology and Toxicology and Chemistry and Biochemistry, University of Arizona Cancer Center, and BIO5 Institute, University of Arizona, Tucson, Arizona, 85721, USA
| |
Collapse
|
40
|
Doelman W, Ligthart NAM, van de Plassche MAT, de Geus MAR, Reinalda L, Isendoorn MME, Filippov DV, van Kasteren SI. Synthesis of Peptides Containing a Combination of Free and 2-trans-Cyclooctene Carbamate Protected Lysine Residues. Chembiochem 2024; 25:e202300786. [PMID: 38126970 DOI: 10.1002/cbic.202300786] [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: 11/22/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 12/23/2023]
Abstract
The allylic trans-cyclooctene (TCO) functionality facilitates powerful control over the spatiotemporal activity of bio-active molecules, enabling precision targeting of druglike and imaging modalities. However, the introduction of this function onto molecules remains chemically challenging, particularly for peptides. Modification with TCOs of this important class of biomolecules remains a challenge, primarily due to the sensitivity of the TCO group to the strong acids typically used in global deprotection during solid phase peptide synthesis. Here, we present a novel synthetic approach to site-selectively introduce TCO-groups in peptides. Our approach utilizes azide groups to mask amine functions, enabling selective introduction of the TCO on a single lysine residue. Staudinger reduction of the azides back to the corresponding amines proceeds without disturbing the sensitive TCO. We show that using our method, we can produce TCO-inactivated antigenic peptides of previously unseen complexity.
Collapse
Affiliation(s)
- W Doelman
- Leiden Institute of Chemistry, Leiden, the Netherlands
| | | | | | - M A R de Geus
- Leiden Institute of Chemistry, Leiden, the Netherlands
| | - L Reinalda
- Leiden Institute of Chemistry, Leiden, the Netherlands
| | | | - D V Filippov
- Leiden Institute of Chemistry, Leiden, the Netherlands
| | | |
Collapse
|
41
|
Dai L, Zhou S, Yang C, Li J, Wang Y, Qin M, Pan L, Zhang D, Qian Z, Wu H. A bioorthogonal cell sorting strategy for isolation of desired cell phenotypes. Chem Commun (Camb) 2024; 60:1916-1919. [PMID: 38259188 DOI: 10.1039/d3cc05604j] [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: 01/24/2024]
Abstract
Here we describe a cost-effective and simplified cell sorting method using tetrazine bioorthogonal chemistry. We successfully isolated SKOV3 cells from complex mixtures, demonstrating efficacy in separating mouse lymphocytes expressing interferon and HeLa cells expressing virally transduced green fluorescent protein post-infection.
Collapse
Affiliation(s)
- Liqun Dai
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Siming Zhou
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Cheng Yang
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Jie Li
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yayue Wang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| | - Meng Qin
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lili Pan
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Dan Zhang
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Zhiyong Qian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Haoxing Wu
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
42
|
Huang W, Gunawardhana N, Zhang Y, Escorihuela J, Laughlin ST. Pyranthiones/Pyrones: "Click and Release" Donors for Subcellular Hydrogen Sulfide Delivery and Labeling. Chemistry 2024; 30:e202303465. [PMID: 37985373 DOI: 10.1002/chem.202303465] [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: 10/20/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
Hydrogen sulfide (H2 S), one of the most important gasotransmitters, plays a critical role in endogenous signaling pathways of many diseases. However, developing H2 S donors with both tunable release kinetics and high release efficiency for subcellular delivery has been challenging. Here, we describe a click and release reaction between pyrone/pyranthiones and bicyclononyne (BCN). This reaction features a release of CO2 /COS with second-order rate constants comparable to Strain-Promoted Azide-Alkyne Cycloaddition reactions (SPAACs). Interestingly, pyranthiones showed enhanced reaction rates compared to their pyrone counterparts. We investigated pyrone biorthogonality and demonstrated their utility in protein labeling applications. Moreover, we synthesized substituted pyranthiones with H2 S release kinetics that can address the range of physiologically relevant H2 S dynamics in cells and achieved quantitative H2 S release efficiency in vitro. Finally, we explored the potential of pyranthiones as H2 S/COS donors for mitochondrial-targeted H2 S delivery in living cells.
Collapse
Affiliation(s)
- Wei Huang
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11790, United States
| | - Nipuni Gunawardhana
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11790, United States
| | - Yunlei Zhang
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11790, United States
| | - Jorge Escorihuela
- Departamento de Química Orgánica, Universitat de València, Avda. Vicente Andrés Estellés s/n, Burjassot, 46100, Valencia, Spain
| | - Scott T Laughlin
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11790, United States
| |
Collapse
|
43
|
Dudchak R, Podolak M, Holota S, Szewczyk-Roszczenko O, Roszczenko P, Bielawska A, Lesyk R, Bielawski K. Click chemistry in the synthesis of antibody-drug conjugates. Bioorg Chem 2024; 143:106982. [PMID: 37995642 DOI: 10.1016/j.bioorg.2023.106982] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/31/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023]
Abstract
Antibody-Drug Conjugates (ADC) are a new class of anticancer therapeutics with immense potential. They have been rapidly advancing in the last two decades. This fast speed of development has become possible due to several new technologies and methods. One of them is Click Chemistry, an approach that was created only two decades ago, but already is actively utilized for bioconjugation, material science and drug discovery. In this review, we researched the impact of Click Chemistry reactions on the synthesis and development of ADCs. The information about the most frequently utilized reactions, such as Michael's addition, Copper-catalyzed azide-alkyne [3+2] cycloaddition (CuAAC), Strain-promoted azide-alkyne [3+2] cycloaddition (SPAAC), oxime bond formation, hydrazine-iso-Pictet-Spengler Ligation (HIPS), Diels-Alder reactions have been summarized. The implementation of thiol-maleimide Click Chemistry reaction in the synthesis of numerous FDA-approved Antibody-Drug Conjugates has been reported. The data amassed in the present review provides better understanding of the importance of Click Chemistry in the synthesis, development and improvement of the Antibody-Drug Conjugates and it will be helpful for further researches related to ADCs.
Collapse
Affiliation(s)
- Rostyslav Dudchak
- Department of Synthesis and Technology of Drugs, Faculty of Pharmacy, Medical University of Bialystok, Jana Kilińskiego 1, Bialystok 15-089, Poland
| | - Magdalena Podolak
- Department of Biotechnology, Faculty of Pharmacy, Medical University of Bialystok, Jana Kilińskiego 1, Bialystok 15-089, Poland
| | - Serhii Holota
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, Lviv 79010, Ukraine
| | - Olga Szewczyk-Roszczenko
- Department of Synthesis and Technology of Drugs, Faculty of Pharmacy, Medical University of Bialystok, Jana Kilińskiego 1, Bialystok 15-089, Poland
| | - Piotr Roszczenko
- Department of Biotechnology, Faculty of Pharmacy, Medical University of Bialystok, Jana Kilińskiego 1, Bialystok 15-089, Poland
| | - Anna Bielawska
- Department of Biotechnology, Faculty of Pharmacy, Medical University of Bialystok, Jana Kilińskiego 1, Bialystok 15-089, Poland
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, Lviv 79010, Ukraine.
| | - Krzysztof Bielawski
- Department of Synthesis and Technology of Drugs, Faculty of Pharmacy, Medical University of Bialystok, Jana Kilińskiego 1, Bialystok 15-089, Poland
| |
Collapse
|
44
|
Segawa S, He X, Tang BZ. Metal-free click and bioorthogonal reactions of aggregation-induced emission probes for lighting up living systems. LUMINESCENCE 2024; 39:e4619. [PMID: 37987236 DOI: 10.1002/bio.4619] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 11/22/2023]
Abstract
In 2002, two transformative research paradigms emerged: 'click chemistry' and 'aggregation-induced emission (AIE),' both leaving significant impacts on early 21st-century academia. Click chemistry, which describes the straightforward and reliable reactions for linking two building blocks, has simplified complex molecular syntheses and functionalization, propelling advancements in polymer, material, and life science. In particular, nontoxic, metal-free click reactions involving abiotic functional groups have matured into bioorthogonal reactions. These are organic ligations capable of selective and efficient operations even in congested living systems, therefore enabling in vitro to in vivo biomolecular labelling. Concurrently, AIE, a fluorogenic phenomenon of twisted π-conjugated compounds upon aggregation, has offered profound insight into solid-state photophysics and promoted the creation of aggregate materials. The inherent fluorogenicity and aggregate-emission properties of AIE luminogens have found extensive application in biological imaging, characterized by their high-contrast and photostable fluorescent signals. As such, the convergence of these two domains to yield efficient labelling with excellent fluorescence images is an anticipated progression in recent life science research. In this review, we intend to showcase the synergetic applications of AIE probes and metal-free click or bioorthogonal reactions, highlighting both the achievements and the unexplored avenues in this promising field.
Collapse
Affiliation(s)
- Shinsuke Segawa
- Department of Chemical and Biological Engineering, School of Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Xuewen He
- The Key Lab of Health Chemistry and Molecular Diagnosis of Suzhou, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu, China
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction and Institute for Advanced Study, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| |
Collapse
|
45
|
Min Q, Ji X. Bioorthogonal Bond Cleavage Chemistry for On-demand Prodrug Activation: Opportunities and Challenges. J Med Chem 2023; 66:16546-16567. [PMID: 38085596 DOI: 10.1021/acs.jmedchem.3c01459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Time- and space-resolved drug delivery is highly demanded for cancer treatment, which, however, can barely be achieved with a traditional prodrug strategy. In recent years, the prodrug strategy based on a bioorthogonal bond cleavage chemistry has emerged with the advantages of high temporospatial resolution over drug activation and homogeneous activation irrespective of individual heterogeneity. In the past five years, tremendous progress has been witnessed in this field with one such bioorthogonal prodrug entering Phase II clinical trials. This Perspective aims to highlight these new advances (2019-2023) and critically discuss their pros and cons. In addition, the remaining challenges and potential strategic directions for future progress will also be included.
Collapse
Affiliation(s)
- Qingqiang Min
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| | - Xingyue Ji
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| |
Collapse
|
46
|
Xu M, Ma X, Pigga JE, Zhang H, Wang S, Zhao W, Deng H, Wu AM, Liu R, Wu Z, Fox JM, Li Z. Development of 18F-Labeled hydrophilic trans-cyclooctene as a bioorthogonal tool for PET probe construction. Chem Commun (Camb) 2023; 59:14387-14390. [PMID: 37877355 PMCID: PMC10785124 DOI: 10.1039/d3cc04212j] [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] [Indexed: 10/26/2023]
Abstract
We report the development of a hydrophilic 18F-labeled a-TCO derivative [18F]3 (log P = 0.28) through a readily available precursor and a single-step radiofluorination reaction (RCY up to 52%). We demonstrated that [18F]3 can be used to construct not only multiple small molecule/peptide-based PET agents, but protein/diabody-based imaging probes in parallel.
Collapse
Affiliation(s)
- Muyun Xu
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA.
| | - Xinrui Ma
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA.
| | - Jessica E Pigga
- Department of Chemistry, the University of Delaware, Newark, Delaware, 19716, USA.
| | - He Zhang
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA.
| | - Shuli Wang
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA.
| | - Weiling Zhao
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA.
| | - Huaifu Deng
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA.
| | - Anna M Wu
- Department of Molecular Imaging and Therapy, Beckman Research Institute, City of Hope, Duarte, California, 91010, USA
| | - Rihe Liu
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Zhanhong Wu
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA.
| | - Joseph M Fox
- Department of Chemistry, the University of Delaware, Newark, Delaware, 19716, USA.
| | - Zibo Li
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA.
| |
Collapse
|
47
|
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: 22] [Impact Index Per Article: 11.0] [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
|
48
|
Yan Z, Pan Y, Jiao G, Xu M, Fan D, Hu Z, Wu J, Chen T, Liu M, Bao X, Ke H, Ji X. A Bioorthogonal Decaging Chemistry of N-Oxide and Silylborane for Prodrug Activation both In Vitro and In Vivo. J Am Chem Soc 2023; 145:24698-24706. [PMID: 37933858 DOI: 10.1021/jacs.3c08012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Bioorthogonal decaging chemistry with both fast kinetics and high efficiency is highly demanded for in vivo applications but remains very sporadic. Herein, we describe a new bioorthogonal decaging chemistry between N-oxide and silylborane. A simple replacement of "C" in boronic acid with "Si" was able to substantially accelerate the N-oxide decaging kinetics by 106 fold (k2: up to 103 M-1 s-1). Moreover, a new N-oxide-masked self-immolative spacer was developed for the traceless release of various payloads upon clicking with silylborane with fast kinetics and high efficiency (>90%). Impressively, one such N-oxide-based self-assembled bioorthogonal nano-prodrug in combination with silylborane led to significantly enhanced tumor suppression effects as compared to the parent drug in a 4T1 mouse breast tumor model. In aggregate, this new bioorthogonal click-and-release chemistry is featured with fast kinetics and high efficiency and is perceived to find widespread applications in chemical biology and drug delivery.
Collapse
Affiliation(s)
- Zhicheng Yan
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215021, China
| | - Yiyao Pan
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215021, China
| | - Guofeng Jiao
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215021, China
| | - Mengyu Xu
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu 215123, China
| | - Dongguang Fan
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215021, China
| | - Ziwei Hu
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215021, China
| | - Jiarui Wu
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215021, China
| | - Tao Chen
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215021, China
| | - Miao Liu
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215021, China
| | - Xiaoguang Bao
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu 215123, China
| | - Hengte Ke
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215021, China
| | - Xingyue Ji
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215021, China
| |
Collapse
|
49
|
Fu Q, Shen S, Sun P, Gu Z, Bai Y, Wang X, Liu Z. Bioorthogonal chemistry for prodrug activation in vivo. Chem Soc Rev 2023; 52:7737-7772. [PMID: 37905601 DOI: 10.1039/d2cs00889k] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Prodrugs have emerged as a major strategy for addressing clinical challenges by improving drug pharmacokinetics, reducing toxicity, and enhancing treatment efficacy. The emergence of new bioorthogonal chemistry has greatly facilitated the development of prodrug strategies, enabling their activation through chemical and physical stimuli. This "on-demand" activation using bioorthogonal chemistry has revolutionized the research and development of prodrugs. Consequently, prodrug activation has garnered significant attention and emerged as an exciting field of translational research. This review summarizes the latest advancements in prodrug activation by utilizing bioorthogonal chemistry and mainly focuses on the activation of small-molecule prodrugs and antibody-drug conjugates. In addition, this review also discusses the opportunities and challenges of translating these advancements into clinical practice.
Collapse
Affiliation(s)
- Qunfeng Fu
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
- Changping Laboratory, Beijing 102206, China
| | - Siyong Shen
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Pengwei Sun
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Zhi Gu
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Yifei Bai
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Xianglin Wang
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Zhibo Liu
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
- Changping Laboratory, Beijing 102206, China
- Peking University-Tsinghua University Center for Life Sciences, Peking University, Beijing 100871, China
- Key Laboratory of Carcinogenesis and Translational Research of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
50
|
Li Y, Ling Y, Loehr MO, Chaabane S, Cheng OW, Zhao K, Wu C, Büscher M, Weber J, Stomakhine D, Munker M, Pientka R, Christ SB, Dobbelstein M, Luedtke NW. DNA templated Click Chemistry via 5-vinyl-2'-deoxyuridine and an acridine-tetrazine conjugate induces DNA damage and apoptosis in cancer cells. Life Sci 2023; 330:122000. [PMID: 37541577 DOI: 10.1016/j.lfs.2023.122000] [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: 04/02/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023]
Abstract
AIMS Click Chemistry is providing valuable tools to biomedical research, but its direct use in therapies remains nearly unexplored. For cancer treatment, nucleoside analogues (NA) such as 5-vinyl-2'-deoxyuridine (VdU) can be metabolically incorporated into cancer cell DNA and subsequently "clicked" to form a toxic product. The inverse electron-demand Diels-Alder (IEDDA) reaction between VdU and an acridine-tetrazine conjugate (PINK) has previously been used to label cell nuclei of cultured cells. Here, we report tandem usage of VdU and PINK to induce cytotoxicity. MAIN METHODS Cell lines were subsequently treated with VdU and PINK, and cell viability was measured via well confluency and 3D tumor spheroid assays. DNA damage and apoptosis were evaluated using Western Blotting and cell cycle analysis by flow cytometry. Double stranded DNA break (DSB) formation was measured using the comet assay. Apoptosis was assessed by fluorescent detection of externalized phosphatidylserine residues. KEY FINDINGS We report that the combination of VdU and PINK synergistically induces cytotoxicity in cultured human cells. The combination of VdU and PINK strongly reduced cell viability in 2D and 3D cultured cancer cells. Mechanistically, the compounds induced DNA damage through DSB formation, which leads to S-phase accumulation and apoptosis. SIGNIFICANCE The combination of VdU and PINK represents a novel and promising DNA-templated "click" approach for cancer treatment via selective induction of DNA damage.
Collapse
Affiliation(s)
- Yizhu Li
- 2(nd) Medical Clinic, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22 81675 Munich, Germany.
| | - Yurong Ling
- 2(nd) Medical Clinic, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22 81675 Munich, Germany
| | - Morten O Loehr
- Department of Chemistry, McGill University, 845 Sherbrooke St W, Montreal, Quebec H3A 0G4, Canada
| | - Sabrina Chaabane
- 2(nd) Medical Clinic, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22 81675 Munich, Germany
| | - Oh Wan Cheng
- 2(nd) Medical Clinic, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22 81675 Munich, Germany
| | - Kaifeng Zhao
- Department of Chemistry, McGill University, 845 Sherbrooke St W, Montreal, Quebec H3A 0G4, Canada
| | - Chao Wu
- 2(nd) Medical Clinic, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22 81675 Munich, Germany
| | - Moritz Büscher
- 2(nd) Medical Clinic, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22 81675 Munich, Germany
| | - Jana Weber
- 2(nd) Medical Clinic, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22 81675 Munich, Germany
| | - Daria Stomakhine
- Department of Chemistry, McGill University, 845 Sherbrooke St W, Montreal, Quebec H3A 0G4, Canada
| | - Marina Munker
- 2(nd) Medical Clinic, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22 81675 Munich, Germany
| | - Ronja Pientka
- 2(nd) Medical Clinic, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22 81675 Munich, Germany
| | - Sarah B Christ
- 2(nd) Medical Clinic, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22 81675 Munich, Germany
| | - Matthias Dobbelstein
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, 37077 Göttingen, Germany
| | - Nathan W Luedtke
- Department of Chemistry, McGill University, 845 Sherbrooke St W, Montreal, Quebec H3A 0G4, Canada; Department of Pharmacology and Therapeutics, McGill University, Montréal H3G 1Y6, Canada
| |
Collapse
|