DOI:
10.1039/D5CB00186B
(Paper)
RSC Chem. Biol., 2025, Advance Article
Chemically inducible antisense oligonucleotides for cell-specific gene silencing
Received
18th July 2025
, Accepted 25th September 2025
First published on 9th October 2025
Abstract
Cell-specific control of the function of antisense oligonucleotides (ASOs) is highly desirable for precise gene therapy while minimizing adverse effects in normal cells. Herein, we report a novel class of chemically inducible ASOs (iASOs) that achieve tumor-cell-selective gene silencing through hydrogen peroxide (H2O2)-triggered activation. Through post-synthetic incorporation of phenylboronic acid (BO) caging groups at the backbone positions, we developed iASOs that remain functionally inactive until the H2O2-triggered removal of the BO groups caused activation. Using EGFP as a reporter system, we demonstrated that the optimal BO-modified iASO exhibited slight gene silencing activity in normal cells but achieved >80% knockdown of the target mRNA in tumor cells. The BO-modified iASO was further applied to target the endogenous Bcl2 gene, demonstrating its ability for controlling gene silencing and inducing cell death. This study establishes a simple and effective platform for conditional gene regulation and the development of cell-specific ASO therapeutics.
Introduction
Antisense oligonucleotides (ASOs) have emerged as highly promising therapeutic agents for regulating gene expression.1–7 ASOs are single-stranded synthetic nucleic acids that specifically hybridize with their target mRNA through Watson–Crick base pairing, thereby modulating gene expression through several mechanisms, including ribonuclease H-mediated mRNA degradation, RNA splicing modulation and the steric inhibition of mRNA translation.8–12 Owing to their high programmability, ASOs offer exceptional targeting versatility that allow for the rational design of almost any mRNA of interest. Currently, several ASO drugs, such as Inotersen,13 Volanesorsen,14,15 and Nusinersen,16,17 have been approved, and an increasing number of drug candidates targeting different diseases have also entered clinical trials.18,19 Despite their promise, a critical pharmacological limitation of ASOs is their constitutive activity; that is, upon cellular uptake, therapeutic ASOs induce rapid and sustained gene silencing without precise control of their functions. This “always-on” characteristic raises the risk of off-target effects and systemic toxicity, particularly in non-target tissues where prolonged ASO activity may result in adverse effects.
To address this issue, several strategies that enable the precise control of ASO function have been developed. For example, aptamer-mediated conformational switches have been designed that allow for the regulation of gene expression through exposure of the antisense sequence, triggered by ligand binding.20,21 An alternative approach is constructing chemically caged ASOs through the site-specific incorporation of stimuli-responsive protecting groups at the conserved positions of ASOs. These engineered ASOs would remain inactive under physiological conditions until specific stimuli trigger decaging, thereby activating their functions. Photoactivated ASOs have been developed by incorporating photocleavable groups into the bases and phosphodiester linkages or by constructing cyclized ASOs that inhibit binding of mRNA to the target.22–26 Although these strategies enable the spatiotemporal control of the functions of ASO for conditional gene silencing in cells, their biological applications are largely limited by the requirement for short-wavelength irradiation, which causes poor tissue penetration and potential phototoxicity.27,28 Alternative strategies involving small-molecule-activated ASOs have demonstrated efficient deprotection via the Staudinger reduction, enabling controlled gene silencing in live cells and zebrafish models.29,30 However, the dependence on potentially toxic phosphine-based triggers raises significant safety concerns for clinical applications.31 More recently, endogenous stimulus-responsive ASO systems, utilizing either chemically modified nucleosides or circular ASOs with cleavable linkers, have been developed for cell-specific gene regulation.32–35 For example, Obika et al. designed a series of boronated nucleosides for constructing a H2O2-activated ASO via a solid-phase synthesis.32 Despite their biological relevance, these approaches often involve complex syntheses that may hinder their practical implementation. Therefore, a simple, robust, and effective strategy for achieving cell-selective control of ASO functions remains highly desirable.
In this study, we present a chemically inducible ASO (iASO) design that relies on the site-specific incorporation of caging groups at the backbone positions of ASOs, allowing for the steric hindrance of mRNA hybridization and utilization of endogenous biomolecule-triggered decaging to restore functionality (Scheme 1). As a proof of concept, we selected H2O2 as a model stimulus. H2O2 is a metabolic by-product with elevated levels in tumor cells,36 making it a promising trigger for regulating the function of various molecules, such as small molecules, proteins and nucleic acids.37–45 We designed a series of phenylboronic acid (BO)-modified ASOs via effective phosphorothioate (PS)-bromide chemistry,46 and subsequently identified an iASO with a relatively low basal activity leakage. The H2O2-triggered oxidative hydrolysis of BO enabled the effective removal of the caging groups,47 thereby activating the functioning of ASO (Fig. S1). Our results demonstrated that iASOs exhibited remarkably reduced hybridization capacity and enhanced structural stability in complex biological environments. Upon H2O2-triggered activation, these iASOs enabled the conditional downregulation of the targeted gene expression in tumor cells. Using enhanced green fluorescent protein (EGFP) as an optical reporter, minimal off-target effects were observed in normal cells. Furthermore, we applied this strategy to an inducible ASO that targeted Bcl2, an endogenous, anti-apoptotic gene. The H2O2-triggered activation induced remarkable Bcl2 silencing and subsequent tumor cell death, underscoring the therapeutic potential of this approach. Collectively, our findings establish a simple and effective platform for controllable gene silencing with applications in precision tumor therapy.
 |
| | Scheme 1 Schematic of the H2O2-triggered activation of chemically inducible ASO for controlled gene silencing. | |
Results and discussion
Design of H2O2-inducible AGFP@4BO
To demonstrate the feasibility of constructing iASOs through the site-specific incorporation of caging groups at the backbone positions, we selected EGFP as an optical reporter for evaluating the gene-silencing efficiency. Eight ASO candidates targeting EGFP were initially assessed to identify the most effective sequence for gene regulation. HEK293T cells were co-transfected with EGFP plasmids and each ASO candidate individually, followed by analysis using fluorescence confocal microscopy and flow cytometry. The results indicated that the ASO candidates exhibited varying levels of gene silencing efficiency, with AGFP-8 achieving the highest inhibition rate, as determined by the proportion of GFP-positive cells (Fig. S2). Therefore, AGFP-8 was chosen for subsequent experiments.
To engineer a H2O2-activatable iASO, we initially synthesized a phosphorothioate (PS)-modified AGFP-8 containing four evenly spaced PS modifications (designated as AGFP-8@4PS). The PS modifications not only enhanced the stability of the oligonucleotide, but they also provided reactive sites for the subsequent conjugation of the BO group via bromide-PS chemistry, resulting in BO-modified ASOs (designated as AGFP-8@4BO, Fig. 1A). The synthesis of AGFP-8@4BO was characterized using high performance liquid chromatography (HPLC) and mass spectrometry (MS) analyses. The results show that AGFP-8@4BO exhibited a longer retention time (∼14 min) compared to AGFP-8@4PS (∼13 min), consistent with the increased hydrophobicity conferred by the BO groups. Notably, H2O2 treatment restored the retention time to ∼13 min, indicating that the H2O2-triggered removal of the BO groups (Fig. 1B, left column, Fig. S1). The MS spectra confirmed these observations (Fig. 1B, right column). Collectively, these results demonstrate the feasibility of constructing chemically inducible ASOs.
 |
| | Fig. 1 Characterization of the BO-modified iASO. (A) Schematic of the H2O2-triggered activation of iASO. (B) HPLC (left column) and MS (right column) characterization of AGFP-8@4BO and the H2O2-triggered decaging products (300 μM H2O2 and 100 min). The observed molecular weight of AGFP-8@4BO was smaller than the theoretical value, likely due to the formation of the dehydrated species during ESI-MS analysis. (C) Representative gel image of various concentrations used for the H2O2-triggered decaging of AGFP-8@4BO (top panel). Calculated dose-dependent decaging efficiency (bottom panel). (D) Representative gel image of the H2O2-triggered decaging of AGFP-8@4BO at different times (top panel). The calculated time-dependent decaging efficiency (bottom panel). (E) Representative gel image of the H2O2-triggered activation of iASO for target mRNA hybridization. Error bars represent standard deviation from the three parallel experiments. The decaging reactions were performed in 1× Tris-NaCl buffer: 10 mM Tris-HCl, 140 mM NaCl, and pH 7.4. | |
The dose- and time-dependent decaging efficiencies were evaluated. The incubation of AGFP-8@4BO with varying concentrations of H2O2 for 100 min resulted in a progressively increased decaging efficiency, with 300 μM H2O2 achieving ∼99% decaging efficiency (Fig. 1C). Similarly, incubation with 300 μM μH2O2 over different time periods revealed a time-dependent decaging process with a t1/2 of 34.79 min (Fig. 1D). These results confirm that the H2O2-triggered decaging reaction occurred in both dose- and time-dependent manners. The specificity of the H2O2-triggered decaging was also investigated. The result show that upon H2O2 treatment, AGFP-8@4BO exhibited a remarkably increased bond migration rate compared to AGFP-8@4BO, whereas the other oxidizing agents induced minimal effects (Fig. S3). This result demonstrates the good selectivity of the BO-modified ASO for H2O2, consistent with the previous reports that the BO group serves as an H2O2-responsive motif.48
We further investigated the ability of AGFP-8@4BO for modulating hybridization with the target mRNAs (Fig. 1E). The results demonstrated that AGFP-8@4BO significantly inhibited hybridization activity toward mEGFP (lane 5), whereas H2O2 treatment fully restored this activity (lane 6). The DNA melting analysis revealed that AGFP-8@4BO exhibited a ∼10 °C drop in the melting temperature compared to AGFP-8@4PS, suggesting that the incorporation of the BO groups into ASO significantly blocked its hybridization capacity (Fig. S4). Furthermore, the H2O2-activated AGFP-8@4BO showed a recovered melting temperature comparable to that of AGFP-8@4PS. These findings together underscore the potential of AGFP-8@4BO for achieving controllable gene silencing. Moreover, the structural stability of AGFP-8@4BO was further enhanced due to increased steric hindrance conferred by the chemical modifications. As shown in Fig. S5, ∼70% of AGFP-8@4BO remained intact after a 4-h incubation in 10% fetal bovine serum (FBS) solution, which was higher than that of AGFP-8@4PS and AGFP-8.
Encouraged by the results of H2O2-triggered activation of ASO functions, we further assessed the potential of AGFP-8@4BO for controlled gene silencing in live cells, using EGFP as an optical reporter. Prior to conducting experiments, the cytotoxicity of H2O2 was evaluated in HEK293T cells to determine the maximum concentration (maintaining ∼90% cell viability) that could be used for subsequent activation experiments (Fig. S6A). The results indicate that AGFP-8@4PS caused a relatively higher reduction in EGFP expression in HEK293T cells compared to AGFP-8 (Fig. S7), primarily due to the enhanced structural stability provided by the PS modifications. Furthermore, only AGFP-8@4BO had a slight impact on EGFP expression, whereas sequential treatment with AGFP-8@4BO and H2O2 led to a significant decrease in EGFP levels in HEK293T cells (Fig. S7). Notably, only cells transfected with EGFP plasmids exhibited negligible changes in their fluorescence under identical H2O2 treatment conditions (Fig. S6B). These results confirm the H2O2-triggered activation of AGFP-8@4BO for controllable gene silencing in living cells.
Live cell characterization of H2O2 inducible AGFP@4BO
We then optimized the number and sites of chemical modifications to identify the optimal iASO. The results indicate that AGFP-8 containing three evenly spaced BO modifications (designated as AGFP-8@3BO as shown in Fig. 2A, S8A and S8B) exhibited noticeable activity leakage in HEK293T cells, whereas AGFP-8@4BO demonstrated much lower gene silencing activity (Fig. 2B and Fig. S8C), suggesting that at least four BO modifications were required to effectively suppress its function. The influence of modification sites on the gene-silencing performance was also examined. The results revealed that the AGFP-8@4BO variants with four BO modifications at different backbone positions displayed variable inhibition and activation efficiencies (Fig. 2A and Fig. S9). Among these variants, AGFP-8@4BO-2 showed the lowest basal activity leakage prior to H2O2-triggered decaging, and subsequent H2O2-triggered decaging enabled the efficient activation of gene silencing (Fig. 2C, D and Fig. S10). Collectively, these results suggest that AGFP-8@4BO-2 functioned as an optimal iASO for controlled gene regulation in living cells.
 |
| | Fig. 2 H2O2-triggered activation of AGFP-8@4BO for cell-specific gene silencing of exogenous EGFP mRNA. (A) Schematic of the BO modifications on ASOs. Gene silencing efficiency of the iASO candidates with different modification numbers (B) or different modification sites (C), as measured by flow cytometric analysis of GFP expression in HEK293T cells. (D) Confocal imaging of HEK293T cells transfected with AGFP-8@4BO-2, followed by treatment with or without H2O2. (E) Confocal imaging of HEK293T and Ncl-H460 cells transfected with AGFP-8@4BO-2. | |
It has been shown that the overproduction of H2O2 is closely associated with various diseases including cancer, making it a widely utilized stimulus for activating prodrugs in tumor therapy. We subsequently evaluated the ability of AGFP-8@4BO-2 for achieving tumor cell-specific gene regulation. A human lung carcinoma cell line, Ncl-H460, and a normal human embryonic kidney cell line, HEK293T, were selected as model cell lines for comparison. Compared to HEK293T cells co-transfected with EGFP plasmids and AGFP-8@4BO-2, which showed only a modest reduction in EGFP expression (∼20% gene silencing efficiency), cancerous Ncl-H460 cells co- transfected with the same constructs exhibited a substantial decrease in EGFP fluorescence, corresponding to ∼80% gene silencing efficiency (Fig. 2E and Fig. S11A–C). This result was consistent with the intracellular H2O2 levels in these two cell lines, as quantified using a commercially available H2O2 assay kit (Fig. S11D). Collectively, these results demonstrate the H2O2-triggered activation of iASO for tumor cell-specific gene silencing.
Live cell gene silencing of Bcl2 using H2O2 inducible ABcl2@BO
Having demonstrated the efficient gene silencing of the exogenous mRNA, we proceeded to design another iASO that targeted the endogenous mRNA, Bcl2, a key regulator of cell apoptosis. Previous studies have established that Bcl2 plays a critical role in promoting cell survival in most mammalian cells and contributes to chemoresistance in cancer.49 The gene regulation capability of the previously reported Bcl2-targeting ASO, G313950 was first validated (Fig. S12). The BO-modified ABcl2 was then synthesized according to the developed method (designated as ABcl2@BO, Fig. S13A) and characterized by HPLC and MS analyses (Fig. S13C). Additionally, a negative control ABcl2 modified with phenylacetic acid groups, which cannot respond to H2O2 stimulation, was constructed (designated as ABcl2@CC, Fig. S13B and S13C). Consistent with previous observations, ABcl2@BO exhibited significantly reduced hybridization activity (Fig. S13D), which was restored upon H2O2-triggered activation. In contrast, the hybridization capacity of ABcl2@CC remained constitutively suppressed (Fig. S10E).
We then assessed the therapeutic efficacy of ABcl2@BO in human breast cancer MCF-7 cells. Reverse transcription-polymerase chain reaction (RT-PCR) analysis revealed a significant reduction in Bcl2 mRNA expression in cells sequentially treated with ABcl2@BO and H2O2 compared to untreated cells (Fig. 3A), whereas a minimal decrease was observed in the control groups (ABcl2@BO alone, ABcl2@CC and ABcl2@CC + H2O2). Western blot analysis confirmed this result at the protein level (Fig. 3B). These results demonstrate the H2O2-triggered activation of ABcl2@BO for controlled gene silencing. We further investigated the effect of Bcl2 silencing on cell viability. Our data show that the sequential treatment of MCF-7 cells with ABcl2@BO and H2O2 led to a significantly increased release of lactate dehydrogenase (LDH) compared to the control groups (Fig. 3C). Consequently, a substantial reduction in the viable cell count was observed following co-treatment with ABcl2@BO and H2O2 (Fig. 3D). Moreover, the PI/Annexin V-FITC apoptosis assay demonstrated that the H2O2-activated ABcl2@BO induced a higher apoptosis rate than the control groups (Fig. 3E), which aligned with the results from the crystal violet assay (Fig. 3F). Collectively, these results demonstrate that the engineered H2O2-activated iASO enabled precise gene silencing in living cells and exhibited potential for application in cancer gene therapy.
 |
| | Fig. 3 H2O2-triggered activation of ABcl2@BO for controllable gene regulation and antitumor studies in MCF-7 cells. (A) RT-PCR analysis of Bcl2 mRNA expression under different treatments. (B) Western blot analysis of Bcl2 protein expression levels. Cell viability analysis by (C) the LDH release assay and (D) direct cell counting. (E) Cell apoptosis analysis using Annexin V/PI staining. (F) Cell survival evaluation by crystal violet staining. Error bars represent standard deviation from three parallel experiments. Statistical significance was calculated with a two-tailed Student's t-test: ****P < 0.0001. | |
Conclusions
In this study, we developed a H2O2-activatable iASO by site-specifically modifying its backbone with BO caging groups. The resulting iASOs demonstrated not only enhanced metabolic stability, but also the capacity for the precise control of their functions via H2O2-triggered activation. Our results showed that the BO-modified iASOs remained biologically inactive in normal cells, but they effectively silenced EGFP mRNA in tumor cells with elevated H2O2 levels. Furthermore, we demonstrated that the H2O2-activated iASOs targeting the endogenous Bcl2 mRNA significantly induced apoptosis using a lipid nanoparticle system, thereby confirming their therapeutic potential. Therefore, our strategy provides a simple and robust platform for cell-specific gene regulation, underscoring its promise in minimizing off-target effects and advancing precision oncology.
Author contributions
Zhen Xun and Yang Hai contributed equally to this work. Zhen Xun performed the experiments and analysed the data; Yang Hai performed the organic synthesis; Li-Juan Tang discussed and wrote the manuscript; Jian-Hui Jiang discussed the results; Zhenkun Wu designed the project and wrote the manuscript.
Conflicts of interest
There are no conflicts to declare.
Data availability
The data supporting this article, including DNA sequences, gel electrophoresis analysis, confocal images,and flow cytometric analysis, have been included in the supplementary information (SI). See DOI: https://doi.org/10.1039/d5cb00186b.
Acknowledgements
This project is supported by the National Natural Science Foundation of China (22174043 and 22090050), Science and Technology Major Project of Hunan Province (2021SK1020) and Hunan Provincial Science Fund for Distinguished Young Scholars (2024JJ2011). MCF-7, NCI-H460, and HEK293T cell lines were obtained from the Cell Bank of the Chinese Academy of Sciences Committee on Type Culture Collection (Shanghai, China).
Notes and references
- E. Ersöz and D. Demir-Dora, Drug Dev. Res., 2024, 85, e22178 CrossRef PubMed.
- S. T. Crooke, B. F. Baker, R. M. Crooke and X.-H. Liang, Nat. Rev. Drug Discovery, 2021, 20, 427–453 CrossRef CAS PubMed.
- J. C. Means, A. L. Martinez-Bengochea, D. A. Louiselle, J. M. Nemechek, M. Perry, E. G. Farrow, T. S. Pastinen and T. Younger, Nature, 2025, 638, 239–243 CrossRef PubMed.
- B. E. Cook, T. C. Pickel, S. Nag, P. N. Bolduc, R. Beshr, A. F. Morén, C. Muste, G. Boscutti, D. Jiang, L. Yuan, P. Datta, P. Ochniewicz, Y. K. Meynaq, S. P. Tang, C. Plisson, M. Amatruda, Q. Zhang, J. M. DuBois, A. Delavari, S. K. Klein, I. Polyak, A. Shoroye, S. Girmay, C. Halldin, L. Martarello, E. A. Peterson and M. Kaliszczak, Sci. Transl. Med., 2025, 17, eadl1732 CrossRef CAS PubMed.
- B. Borges, S. M. Brown, W. J. Chen, M. T. Clarke, A. Herzeg, J. H. Park, J. Ross, L. Kong, M. Denton, A. K. Smith, T. Lum, F. M. Zada, M. Cordero, N. Gupta, S. E. Cook, H. Murray, J. Matson, S. Klein, C. F. Bennett, A. R. Krainer, T. C. MacKenzie and C. J. Sumner, Sci. Transl. Med., 2025, 17, eadv4656 CrossRef CAS PubMed.
- Z. T. McEachin, M. Chung, S. A. Stratton, C. Han, W. J. Kim, U. Sheth, E. V. Thomas, E. Issenberg, T. Kamra, P. Merino, Y. Levites, N. Raj, E. B. Dammer, D. M. Duong, L. Ping, A. Shantaraman, A. N. Trautwig, J. Gadhavi, E. Assefa, M. Gearing, K. M. Kelly, S. F. Roemer, M. D. Ture, S. Asress, T. Kukar, C. Fournier, D. W. Dickson, L. Petrucelli, T. E. Golde, B. Oskarsson, T. F. Gendron, N. T. Seyfried and J. D. Glass, Cell, 2025, 188, 1–12 CrossRef PubMed.
- M. Egli and M. Manoharan, Nucleic Acids Res., 2023, 51, 2529–2573 CrossRef CAS PubMed.
- J. Hörberg, A. Carlesso and A. Reymer, Mol. Ther. Nucleic Acids, 2024, 35, 102351 CrossRef PubMed.
- S. T. Crooke, Nucleic Acid Ther., 2017, 27, 70–77 CrossRef CAS PubMed.
- D. Collotta, I. Bertocchi, E. Chiapello and M. Collino, Front. Pharmacol., 2023, 14, 1304342 CrossRef CAS PubMed.
- C. F. Bennett and E. E. Swayze, Annu. Rev. Pharmacol. Toxicol., 2010, 50, 259–293 CrossRef CAS PubMed.
- C. F. Bennett, A. R. Krainer and D. W. Cleveland, Annu. Rev. Neurosci., 2019, 42, 385–406 CrossRef CAS PubMed.
- S. J. Keam, AdisInsight Rep., 2018, 78, 1371–1376 CAS.
- M. C. Izar and F. A. H. Fonseca, Curr. Atheroscler. Rep., 2025, 17, 51 CrossRef PubMed.
- R. Hanssen and I. G. Berthold, Curr. Pharmacol. Rep., 2017, 3, 458–468 CrossRef CAS.
- M. J. A. Wood, K. Talbot and M. Bowerman, Hum. Mol. Genet., 2017, 26, 151–159 CrossRef PubMed.
- B. T. Darras, C. A. Chiriboga, S. T. Iannaccone, K. J. Swoboda, J. Montes, L. Mignon, S. Xia, C. F. Bennett, K. M. Bishop, J. M. Shefner, A. M. Green, P. Sun, I. Bhan, S. Gheuens, E. Schneider, W. Farwell and D. C. D. Vivo, Neurology, 2019, 92, 2492–2506 CrossRef PubMed.
- Y. Miao, C. Fu, Z. Yu, L. Yu, Y. Tang and M. Wei, Nucleic Acid Ther., 2023, 32, 83–94 Search PubMed.
- H. Takakusa, N. Iwazaki, M. Nishikawa, T. Yoshida, S. Obika and T. Inoue, Acta Pharm Sin B, 2024, 14, 3802–3817 CrossRef PubMed.
- S. Stangherlin, N. Lui, J. H. Lee and J. W. Liu, Trends Anal. Chem., 2025, 191, 118349 CrossRef CAS.
- T. S. Bayer and C. D. Smolke, Nat. Biotechnol., 2005, 23, 337–343 CrossRef CAS PubMed.
- D. Hartmann and M. J. Booth, Commun. Chem., 2023, 6, 59 CrossRef CAS PubMed.
- D. Hartmann and M. J. Booth, Chem. Commun., 2023, 59, 5685–5688 RSC.
- X. Tang, M. Su, L. Yu, C. Lv, J. Wang and Z. Li, Nucleic Acids Res., 2010, 38, 3848–3855 CrossRef CAS.
- L. Yang, H. B. Kim, J. Y. Sul, S. B. Yeldell, J. H. Eberwine and I. J. Dmochowski, ChemBioChem, 2018, 19, 1250–1254 CrossRef CAS PubMed.
- X. Tang and I. J. Dmochowski, Nat. Protoc., 2007, 1, 3041–3048 CrossRef.
- D. Broadwater, M. Bates, M. Jayaram, M. Young, J. He, A. L. Raithel, T. W. Hamann, W. Zhang, B. Borhan, R. R. Lunt and S. Y. Lunt, Sci. Rep., 2019, 9, 15288 CrossRef.
- J. Zhao, H. Chu, Y. Zhao, Y. Lu and L. Li, J. Am. Chem. Soc., 2019, 141, 7056–7062 CrossRef CAS PubMed.
- A. Bardhan, W. Brown, S. Albright, M. Tsang, L. A. Davidson and A. Deiters, Angew. Chem., Int. Ed., 2024, 63, e202318773 CrossRef CAS.
- K. Darrah, J. Wesalo, B. Lukasak, M. Tsang, J. K. Chen and A. Deiters, J. Am. Chem. Soc., 2021, 143, 18665–18671 CrossRef CAS PubMed.
- N. S. Nath, I. Bhattacharya, A. G. Tuck, D. I. Schlipalius and P. R. Ebert, J. Toxicol., 2011, 2011, 494168 Search PubMed.
- S. Mori, K. Morihiro, T. Okuda, Y. Kasahara and S. Obika, Chem. Sci., 2018, 9, 1112–1118 RSC.
- N. Shirakami, Y. Kawaki, S. L. Higashi, A. Shibata, Y. Kitamura, S. A. Hanifah, L. L. Wah and M. Ikeda, Chem. Lett., 2021, 50, 1412–1415 CrossRef.
- X. Zhao, J. Xu, X. Liang, Z. Wang, Y. Zhu, D. Guo, J. Wang, G. Amu, Q. Wang, Z. Yang and X. Tang, J. Med. Chem., 2025, 68, 4466–4476 CrossRef PubMed.
- Z. Wang, X. Fan, G. Mu, X. Zhao, Q. Wang, J. Wang and X. Tang, Mol. Ther. Nucleic Acids, 2023, 33, 548–558 CrossRef PubMed.
- C. Gorrini, I. S. Harris and T. W. Mak, Nat. Rev. Drug Discovery, 2013, 12, 931–947 CrossRef PubMed.
- J. Liu, J. Liang, C. Wu and Y. Zhao, Anal. Chem., 2019, 91, 6902–6909 Search PubMed.
- R. Weinstain, E. N. Savariar, C. N. Felsen and R. Y. Tsien, J. Am. Chem. Soc., 2014, 136, 874–877 CrossRef PubMed.
- M. Wang, S. Sun, C. I. Neufeld, B. P. Ramirez and Q. Xu, Angew. Chem., Int. Ed., 2014, 53, 13444–13448 CrossRef PubMed.
- Z. Wang, J. Yang, G. Qin, C. Zhao, J. Ren and X. Qu, Angew. Chem., Int. Ed., 2022, 61, e202204291 CrossRef PubMed.
- Y. Liu, Y. Shi, L. Yu, Z. Wu and J. H. Jiang, Anal. Chem., 2023, 95, 6490–6495 CrossRef PubMed.
- H. Ji, W. Xiong, K. Zhang, T. Tian and X. Zhou, Chem. – Asian J., 2022, 17, e202200214 CrossRef PubMed.
- C. Gu, L. Xiao, J. Shang, X. Xu, L. He and Y. Xiang, Chem. Sci., 2021, 12, 9934–9945 RSC.
- L. Xiao, C. Gu and Y. Xiang, Angew. Chem., Int. Ed., 2019, 58, 14167–14172 CrossRef PubMed.
- R. Wang, W. He, X. Yi, Z. Wu, X. Chu and J. H. Jiang, J. Am. Chem. Soc., 2023, 145, 17926–17935 CrossRef PubMed.
- J. A. Fidanza, H. Ozaki and L. W. McLaughlin, J. Am. Chem. Soc., 1992, 114, 5509–5517 CrossRef.
- C. Wang, Y. Wang, G. Wang, C. Huang and N. Jia, Anal. Chim. Acta, 2020, 1097, 230–237 CrossRef CAS PubMed.
- A. R. Lippert, G. C. V. Bittner and C. J. Chang, Acc. Chem. Res., 2011, 44, 793–804 CrossRef CAS PubMed.
- S. Qian, Z. Wei, W. Yang, J. Huang, Y. Yang and J. Wang, Front Oncol., 2022, 12, 985363 CrossRef CAS PubMed.
- X. Pan, L. Chen, S. Liu, X. Yang, J. Gao and R. J. Lee, Mol. Pharm., 2008, 1, 211–220 Search PubMed.
Footnote |
| † These authors contributed equally to this work. |
|
| This journal is © The Royal Society of Chemistry 2025 |
Click here to see how this site uses Cookies. View our privacy policy here.