The Future of Bioorthogonal ChemistryClick to copy article linkArticle link copied!
- Neal K. Devaraj*Neal K. Devaraj*E-mail: [email protected]Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093, United StatesMore by Neal K. Devaraj
Abstract
Bioorthogonal reactions have found widespread use in applications ranging from glycan engineering to in vivo imaging. Researchers have devised numerous reactions that can be predictably performed in a biological setting. Depending on the requirements of the intended application, one or more reactions from the available toolkit can be readily deployed. As an increasing number of investigators explore and apply chemical reactions in living systems, it is clear that there are a myriad of ways in which the field may advance. This article presents an outlook on the future of bioorthogonal chemistry. I discuss currently emerging opportunities and speculate on how bioorthogonal reactions might be applied in research and translational settings. I also outline hurdles that must be cleared if progress toward these goals is to be made. Given the incredible past successes of bioorthogonal chemistry and the rapid pace of innovations in the field, the future is undoubtedly very bright.
Synopsis
Bioorthogonal reactions have found widespread use in chemical biology. This article gives a brief outlook on the future of the field, outlining emerging areas and key challenges to overcome.
Introduction
The Development of New Bioorthogonal Reactions
Assembling Bioactive Compounds for in Situ Drug Delivery
Bioorthogonal Uncaging Reactions
Bioorthogonal Reactions that Lead to Natural Linkages
In Situ Formation of Bioorthogonal Handles
In Vivo Chemistry
Conclusions
Acknowledgments
N.K.D. acknowledges the University of California, San Diego, National Institutes of Health, Department of Defense, and the Camille and Henry Dreyfus foundation for supporting research related to bioorthogonal reactions.
References
This article references 70 other publications.
- 1Hang, H. C.; Yu, C.; Kato, D. L.; Bertozzi, C. R. A metabolic labeling approach toward proteomic analysis of mucin-type O-linked glycosylation. Proc. Natl. Acad. Sci. U. S. A. 2003, 100, 14846– 14851, DOI: 10.1073/pnas.2335201100Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXpvFaqsbk%253D&md5=225b7d0f94f9684e4595c68b725d3425A metabolic labeling approach toward proteomic analysis of mucin-type O-linked glycosylationHang, Howard C.; Yu, Chong; Kato, Darryl L.; Bertozzi, Carolyn R.Proceedings of the National Academy of Sciences of the United States of America (2003), 100 (25), 14846-14851CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Mucin-type O-linked glycoproteins are involved in a variety of biol. interactions in higher eukaryotes. The biosynthesis of these glycoproteins is initiated by a family of polypeptide N-acetyl-α-galactosaminyltransferases (ppGalNAcTs) that modify proteins in the secretory pathway. The lack of a defined consensus sequence for the ppGalNAcTs makes the prediction of mucin-type O-linked glycosylation difficult based on primary sequence alone. Herein we present a method for labeling mucin-type O-linked glycoproteins with a unique chem. tag, the azide, which permits their selective covalent modification from complex cell lysates. From a panel of synthetic derivs., we identified an azido GalNAc analog (N-azidoacetylgalactosamine, GalNAz) that is metabolized by numerous cell types and installed on mucin-type O-linked glycoproteins by the ppGalNAcTs. The azide serves as a bioorthogonal chem. handle for selective modification with biochem. or biophys. probes using the Staudinger ligation. The approach was validated by labeling a recombinant glycoprotein that is known to possess O-linked glycans with GalNAz. In addn., GalNAz efficiently labeled mucin-type O-linked glycoproteins expressed at endogenous levels. The ability to label mucin-type O-linked glycoproteins with chem. tags should facilitate their identification by proteomic strategies.
- 2Bertozzi, C. R. A decade of bioorthogonal chemistry. Acc. Chem. Res. 2011, 44, 651– 653, DOI: 10.1021/ar200193fGoogle Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtFOitrvN&md5=d864c8ac9684dfd32f6c352669d49fdbA Decade of Bioorthogonal ChemistryBertozzi, Carolyn R.Accounts of Chemical Research (2011), 44 (9), 651-653CODEN: ACHRE4; ISSN:0001-4842. (American Chemical Society)There is no expanded citation for this reference.
- 3Sletten, E. M.; Bertozzi, C. R. Bioorthogonal chemistry: fishing for selectivity in a sea of functionality. Angew. Chem., Int. Ed. 2009, 48, 6974– 6998, DOI: 10.1002/anie.200900942Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtV2ltrvK&md5=993199cb4deabc186848bae6fe81f605Bioorthogonal Chemistry: Fishing for Selectivity in a Sea of FunctionalitySletten, Ellen M.; Bertozzi, Carolyn R.Angewandte Chemie, International Edition (2009), 48 (38), 6974-6998CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. The study of biomols. in their native environments is a challenging task because of the vast complexity of cellular systems. Technologies developed in the last few years for the selective modification of biol. species in living systems have yielded new insights into cellular processes. Key to these new techniques are bioorthogonal chem. reactions, whose components must react rapidly and selectively with each other under physiol. conditions in the presence of the plethora of functionality necessary to sustain life. Herein the authors describe the bioorthogonal chem. reactions developed to date and how they can be used to study biomols.
- 4Baskin, J. M.; Prescher, J. A.; Laughlin, S. T.; Agard, N. J.; Chang, P. V.; Miller, I. A.; Lo, A.; Codelli, J. A.; Bertozzi, C. R. Copper-free click chemistry for dynamic in vivo imaging. Proc. Natl. Acad. Sci. U. S. A. 2007, 104, 16793– 16797, DOI: 10.1073/pnas.0707090104Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXht1KgtLvN&md5=7ea429c09b86509ff7f3960a95f11276Copper-free click chemistry for dynamic in vivo imagingBaskin, Jeremy M.; Prescher, Jennifer A.; Laughlin, Scott T.; Agard, Nicholas J.; Chang, Pamela V.; Miller, Isaac A.; Lo, Anderson; Codelli, Julian A.; Bertozzi, Carolyn R.Proceedings of the National Academy of Sciences of the United States of America (2007), 104 (43), 16793-16797CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Dynamic imaging of proteins in live cells is routinely performed by using genetically encoded reporters, an approach that cannot be extended to other classes of biomols. such as glycans and lipids. Here, the authors report a Cu-free variant of click chem. that can label these biomols. rapidly and selectively in living systems, overcoming the intrinsic toxicity of the canonical Cu-catalyzed reaction. The crit. reagent, a substituted cyclooctyne, possesses ring strain and electron-withdrawing fluorine substituents that together promote the [3+2] dipolar cycloaddn. with azides installed metabolically into biomols. This Cu-free click reaction possesses comparable kinetics to the Cu-catalyzed reaction and proceeds within minutes on live cells with no apparent toxicity. With this technique, the authors studied the dynamics of glycan trafficking and identified a population of sialoglycoconjugates with unexpectedly rapid internalization kinetics.
- 5Laughlin, S. T.; Baskin, J. M.; Amacher, S. L.; Bertozzi, C. R. In vivo imaging of membrane-associated glycans in developing zebrafish. Science 2008, 320, 664– 667, DOI: 10.1126/science.1155106Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXltFyis78%253D&md5=2f0a4dbef1ef18bba45013e3847387baIn Vivo Imaging of Membrane-Associated Glycans in Developing ZebrafishLaughlin, Scott T.; Baskin, Jeremy M.; Amacher, Sharon L.; Bertozzi, Carolyn R.Science (Washington, DC, United States) (2008), 320 (5876), 664-667CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Glycans are attractive targets for mol. imaging but have been inaccessible because of their incompatibility with genetically encoded reporters. We demonstrated the noninvasive imaging of glycans in live developing zebrafish, using a chem. reporter strategy. Zebrafish embryos were treated with an unnatural sugar to metabolically label their cell-surface glycans with azides. Subsequently, the embryos were reacted with fluorophore conjugates by means of copper-free click chem., enabling the visualization of glycans in vivo at subcellular resoln. during development. At 60 h after fertilization, we obsd. an increase in de novo glycan biosynthesis in the jaw region, pectoral fins, and olfactory organs. Using a multicolor detection strategy, we performed a spatiotemporal anal. of glycan expression and trafficking and identified patterns that would be undetectable with conventional mol. imaging approaches.
- 6Agarwal, P.; Beahm, B. J.; Shieh, P.; Bertozzi, C. R. Systemic Fluorescence Imaging of Zebrafish Glycans with Bioorthogonal Chemistry. Angew. Chem., Int. Ed. 2015, 54, 11504– 11510, DOI: 10.1002/anie.201504249Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht1Kiu7vL&md5=454dc0471cc02051439df43c3fe4d55cSystemic Fluorescence Imaging of Zebrafish Glycans with Bioorthogonal ChemistryAgarwal, Paresh; Beahm, Brendan J.; Shieh, Peyton; Bertozzi, Carolyn R.Angewandte Chemie, International Edition (2015), 54 (39), 11504-11510CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Vertebrate glycans constitute a large, important, and dynamic set of post-translational modifications that are notoriously difficult to manipulate and image. Although the chem. reporter strategy has been used in conjunction with bioorthogonal chem. to image the external glycosylation state of live zebrafish and detect tumor-assocd. glycans in mice, the ability to image glycans systemically within a live organism has remained elusive. Here, the authors report a method that combines the metabolic incorporation of a cyclooctyne-functionalized sialic acid deriv. with a ligation reaction of a fluorogenic tetrazine, allowing for the imaging of sialylated glycoconjugates within live zebrafish embryos.
- 7Oller-Salvia, B.; Kym, G.; Chin, J. W. Rapid and Efficient Generation of Stable Antibody-Drug Conjugates via an Encoded Cyclopropene and an Inverse-Electron-Demand Diels-Alder Reaction. Angew. Chem., Int. Ed. 2018, 57, 2831– 2834, DOI: 10.1002/anie.201712370Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXis1WjtLg%253D&md5=f2b16b8bdc875d4d0de3b32e649c2e67Rapid and Efficient Generation of Stable Antibody-Drug Conjugates via an Encoded Cyclopropene and an Inverse-Electron-Demand Diels-Alder ReactionOller-Salvia, Benjami; Kym, Gene; Chin, Jason W.Angewandte Chemie, International Edition (2018), 57 (11), 2831-2834CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Homogeneous antibody-drug conjugates (ADCs), generated by site-specific toxin linkage, show improved therapeutic indexes with respect to traditional ADCs. However, current methods to produce site-specific conjugates suffer from low protein expression, slow reaction kinetics, and low yields, or are limited to particular conjugation sites. Here we describe high yielding expression systems that efficiently incorporate a cyclopropene deriv. of lysine (CypK) into antibodies through genetic-code expansion. We express trastuzumab bearing CypK and conjugate tetrazine derivs. to the antibody. We show that the dihydropyridazine linkage resulting from the conjugation reaction is stable in serum, and generate an ADC bearing monomethyl auristatin E that selectively kills cells expressing a high level of HER2. Our results demonstrate that CypK is a minimal bioorthogonal handle for the rapid prodn. of stable therapeutic protein conjugates.
- 8Devaraj, N. K.; Thurber, G. M.; Keliher, E. J.; Marinelli, B.; Weissleder, R. Reactive polymer enables efficient in vivo bioorthogonal chemistry. Proc. Natl. Acad. Sci. U. S. A. 2012, 109, 4762– 4767, DOI: 10.1073/pnas.1113466109Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XltF2msbs%253D&md5=1e747e0637e4f98c48d2204321b8c4efReactive polymer enables efficient in vivo bioorthogonal chemistryDevaraj, Neal K.; Thurber, Greg M.; Keliher, Edmund J.; Marinelli, Brett; Weissleder, RalphProceedings of the National Academy of Sciences of the United States of America (2012), 109 (13), 4762-4767, S4762/1-S4762/2CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)There has been intense interest in the development of selective bioorthogonal reactions or "click" chem. that can proceed in live animals. Until now however, most reactions still require vast surpluses of reactants because of steep temporal and spatial concn. gradients. Using computational modeling and design of pharmacokinetically optimized reactants, we have developed a predictable method for efficient in vivo click reactions. Specifically, we show that polymer modified tetrazines (PMT) are a key enabler for in vivo bioorthogonal chem. based on the very fast and catalyst-free [4 + 2] tetrazine/trans-cyclooctene cycloaddn. Using fluorescent PMT for cellular resoln. and 18F labeled PMT for whole animal imaging, we show that cancer cell epitopes can be easily reacted in vivo. This generic strategy should help guide the design of future chemistries and find widespread use for different in vivo bioorthogonal applications, particularly in the biomedical sciences.
- 9Antonow, D. Fragment-based approaches and the prospect of fragmented prodrugs. Drug Discovery Today 2010, 15, 801– 803, DOI: 10.1016/j.drudis.2010.08.015Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3cfot1GhtA%253D%253D&md5=af44714f2d9ed7711cede8a917f2d071Fragment-based approaches and the prospect of fragmented prodrugsAntonow DyeisonDrug discovery today (2010), 15 (19-20), 801-3 ISSN:.There is no expanded citation for this reference.
- 10Zengeya, T. T.; Garlick, J. M.; Kulkarni, R. A.; Miley, M.; Roberts, A. M.; Yang, Y.; Crooks, D. R.; Sourbier, C.; Linehan, W. M.; Meier, J. L. Co-opting a Bioorthogonal Reaction for Oncometabolite Detection. J. Am. Chem. Soc. 2016, 138, 15813– 15816, DOI: 10.1021/jacs.6b09706Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvVygsr7N&md5=1476ba65466917dffe27c6e137f62c1cCo-opting a Bioorthogonal Reaction for Oncometabolite DetectionZengeya, Thomas T.; Garlick, Julie M.; Kulkarni, Rhushikesh A.; Miley, Mikayla; Roberts, Allison M.; Yang, Youfeng; Crooks, Daniel R.; Sourbier, Carole; Linehan, W. Marston; Meier, Jordan L.Journal of the American Chemical Society (2016), 138 (49), 15813-15816CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Dysregulated metab. is a hallmark of many diseases, including cancer. Methods to fluorescently detect metabolites have the potential to enable new approaches to cancer detection and imaging. However, fluorescent sensing methods for naturally occurring cellular metabolites are relatively unexplored. Here the authors report the development of a chem. approach to detect the oncometabolite fumarate. The authors' strategy exploits a known bioorthogonal reaction, the 1,3-dipolar cycloaddn. of nitrileimines and electron-poor olefins, to detect fumarate via fluorescent pyrazoline cycloadduct formation. The authors demonstrate that hydrazonyl chlorides serve as readily accessible nitrileimine precursors, whose reactivity and spectral properties can be tuned to enable detection of fumarate and other dipolarophile metabolites. Finally, the authors show this reaction can be used to detect enzyme activity changes caused by mutations in fumarate hydratase, which underlie the familial cancer predisposition syndrome hereditary leiomyomatosis and renal cell cancer. The authors' studies define a novel intersection of bioorthogonal chem. and metabolite reactivity that may be harnessed to enable biol. profiling, imaging, and diagnostic applications.
- 11Patterson, D. M.; Prescher, J. A. Orthogonal bioorthogonal chemistries. Curr. Opin. Chem. Biol. 2015, 28, 141– 149, DOI: 10.1016/j.cbpa.2015.07.006Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtlWlsb7J&md5=80c1ef9d29769961cf4f17fbf21d9aa4Orthogonal bioorthogonal chemistriesPatterson, David M.; Prescher, Jennifer A.Current Opinion in Chemical Biology (2015), 28 (), 141-149CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)A review. Bioorthogonal reactions have long been used to examine individual biomols. in living systems. Studies of multi-component networks demand not only reliable bioorthogonal chemistries, but also combinations of reactions that can be used in tandem. Such orthogonal bioorthogonal transformations have been reported in recent years, and these chemistries are enabling new explorations into biol. This article highlights the development of orthogonal bioorthogonal reactions and their application in multi-target imaging and macromol. assembly. Methods to tune and control orthogonal reactivity are also discussed, along with prospects for identifying new classes of compatible reactions.
- 12Yu, J.; Shen, D.; Zhang, H.; Yin, Z. Rapid, Stoichiometric, Site-Specific Modification of Aldehyde-Containing Proteins Using a Tandem Knoevenagel-Intra Michael Addition Reaction. Bioconjugate Chem. 2018, 29, 1016– 1020, DOI: 10.1021/acs.bioconjchem.8b00086Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXislykt70%253D&md5=29f3a6473705c3549232a73aeba3f385Rapid, Stoichiometric, Site-Specific Modification of Aldehyde-Containing Proteins Using a Tandem Knoevenagel-Intra Michael Addition ReactionYu, Jian; Shen, Da; Zhang, Hanjie; Yin, ZhengBioconjugate Chemistry (2018), 29 (4), 1016-1020CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)A site-specific modification of aldehyde-contg. proteins using a tandem Knoevenagel-intra Michael addn. reaction was developed. The reaction featured fast kinetics (50 M-1s-1) and favorable stoichiometry. Various functionalities could be introduced into the protein with little impact on its function and conformation. The reaction was successfully applied in the labeling of living cells.
- 13An, P.; Lewandowski, T. M.; Erbay, T. G.; Liu, P.; Lin, Q. Sterically Shielded, Stabilized Nitrile Imine for Rapid Bioorthogonal Protein Labeling in Live Cells. J. Am. Chem. Soc. 2018, 140, 4860– 4868, DOI: 10.1021/jacs.8b00126Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXls12rsrs%253D&md5=06ffc1977b8a9c476be809a06944438cSterically Shielded, Stabilized Nitrile Imine for Rapid Bioorthogonal Protein Labeling in Live CellsAn, Peng; Lewandowski, Tracey M.; Erbay, Tugce G.; Liu, Peng; Lin, QingJournal of the American Chemical Society (2018), 140 (14), 4860-4868CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)In pursuit of fast bioorthogonal reactions, reactive moieties have been increasingly employed for selective labeling of biomols. in living systems, posing a challenge in attaining reactivity without sacrificing selectivity. To address this challenge, here the authors report a bioinspired strategy in which mol. shape controls the selectivity of a transient, highly reactive nitrile imine dipole. By tuning the shape of structural pendants attached to the ortho position of the N-aryl ring of diaryltetrazoles, precursors of nitrile imines, the authors discovered a sterically shielded nitrile imine that favors the 1,3-dipolar cycloaddn. over the competing nucleophilic addn. The photogenerated nitrile imine exhibits an extraordinarily long half-life of 102 s in aq. medium, owing to its unique mol. shape that hinders the approach of a nucleophile as shown by DFT calcns. The utility of this sterically shielded nitrile imine in rapid (∼1 min) bioorthogonal labeling of glucagon receptor in live mammalian cells was demonstrated.
- 14Matsuo, K.; Nishikawa, Y.; Masuda, M.; Hamachi, I. Live-Cell Protein Sulfonylation Based on Proximity-driven N-Sulfonyl Pyridone Chemistry. Angew. Chem., Int. Ed. 2018, 57, 659– 662, DOI: 10.1002/anie.201707972Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhvFyjsrvN&md5=03016c07f05e88b2836d9746d8e20804Live-Cell Protein Sulfonylation Based on Proximity-driven N-Sulfonyl Pyridone ChemistryMatsuo, Kazuya; Nishikawa, Yuki; Masuda, Marie; Hamachi, ItaruAngewandte Chemie, International Edition (2018), 57 (3), 659-662CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)The development of bioorthogonal approaches for labeling of endogenous proteins under the multimol. crowding conditions of live cells is highly desirable for the anal. and engineering of proteins without using genetic manipulation. N-Sulfonyl pyridone (SP) is reported as a new reactive group for protein sulfonylation. The ligand-directed SP chem. was able to modify not only purified proteins in vitro, but also endogenous ones on the surface of and inside live cells selectively and rapidly, which allowed to convert endogenous proteins to FRET-based biosensors in situ.
- 15Fang, Y. Z.; Zhang, H.; Huang, Z.; Scinto, S. L.; Yang, J. C.; Ende, C. W. A.; Dmitrenko, O.; Johnson, D. S.; Fox, J. M. Photochemical syntheses, transformations, and bioorthogonal chemistry of trans-cycloheptene and sila trans-cycloheptene Ag(I) complexes. Chem. Sci. 2018, 9, 1953– 1963, DOI: 10.1039/C7SC04773HGoogle Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXksFOnsg%253D%253D&md5=0ae7c0fd5e5dc85411d62b7b4be3453aPhotochemical syntheses, transformations, and bioorthogonal chemistry of trans-cycloheptene and sila trans-cycloheptene Ag(I) complexesFang, Yinzhi; Zhang, Han; Huang, Zhen; Scinto, Samuel L.; Yang, Jeffrey C.; am Ende, Christopher W.; Dmitrenko, Olga; Johnson, Douglas S.; Fox, Joseph M.Chemical Science (2018), 9 (7), 1953-1963CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)A photochem. synthesis of AgNO3 complexes of trans-cycloheptene (TCH) and trans-1-sila-4-cycloheptene (Si-TCH) derivs. is described. A low temp. flow photoreactor was designed to enable the synthesis of carbocyclic TCH derivs. due to their thermal sensitivity in the absence of metal coordination. Unlike the free carbocycles, TCH•AgNO3 complexes can be handled at rt and stored for weeks in the freezer (-18 °C). Si-TCH•AgNO3 complexes are esp. robust, and are bench stable for days at rt, and for months in the freezer. X-ray crystallog. was used to characterize a Si-TCH•AgNO3 complex for the first time. With decomplexation of AgNO3 in situ, metal-free TCO and Si-TCH derivs. can engage in a range of cycloaddn. reactions as well as dihydroxylation reactions. Computation was used to predict that Si-TCH would engage in bioorthogonal reactions that are more rapid than the most reactive trans-cyclooctenes. Metal-free Si-TCH derivs. were shown to display good stability in soln., and to engage in the fastest bioorthogonal reaction reported to date (k2 1.14 x 107 M-1s-1 in 9:1 H2O:MeOH). Utility in bioorthogonal protein labeling in live cells is described, including labeling of GFP with an unnatural tetrazine-contg. amino acid. The reactivity and specificity of the Si-TCH reagents with tetrazines in live mammalian cells was also evaluated using the HaloTag platform. The cell labeling expts. show that Si-TCH derivs. are best suited as probe mols. in the cellular environment.
- 16Blackman, M. L.; Royzen, M.; Fox, J. M. Tetrazine ligation: fast bioconjugation based on inverse-electron-demand Diels-Alder reactivity. J. Am. Chem. Soc. 2008, 130, 13518– 13519, DOI: 10.1021/ja8053805Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtFChtrjN&md5=9596082bfdbadaea3cb280a633c34db1Tetrazine Ligation: Fast Bioconjugation Based on Inverse-Electron-Demand Diels-Alder ReactivityBlackman, Melissa L.; Royzen, Maksim; Fox, Joseph M.Journal of the American Chemical Society (2008), 130 (41), 13518-13519CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Described is a bioorthogonal reaction that proceeds with unusually fast reaction rates without need for catalysis: the cycloaddn. of s-tetrazine and trans-cyclooctene derivs. The reactions tolerate a broad range of functionality and proceed in high yield in org. solvents, water, cell media, or cell lysate. The rate of the ligation between trans-cyclooctene and 3,6-di-(2-pyridyl)-s-tetrazine is very rapid (k2 2000 M-1 s-1). This fast reactivity enables protein modification at low concn.
- 17Devaraj, N. K.; Upadhyay, R.; Haun, J. B.; Hilderbrand, S. A.; Weissleder, R. Fast and sensitive pretargeted labeling of cancer cells through a tetrazine/trans-cyclooctene cycloaddition. Angew. Chem., Int. Ed. 2009, 48, 7013– 7016, DOI: 10.1002/anie.200903233Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtV2ltr3P&md5=0caa98e20c8fdd4cf6fec674453b1081Fast and Sensitive Pretargeted Labeling of Cancer Cells through a Tetrazine/trans-Cyclooctene CycloadditionDevaraj, Neal K.; Upadhyay, Rabi; Haun, Jered B.; Hilderbrand, Scott A.; Weissleder, RalphAngewandte Chemie, International Edition (2009), 48 (38), 7013-7016, S7013/1-S7013/6CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)The novel asym. tetrazine I was conjugated to a com. available far-red indocyanine fluorophore, Vivo-Tag 680. This fluorescent tetrazine deriv. was used to image trans-cyclooctene-modified affinity ligands on live cancer cells through a bioorthogonal cycloaddn. with a reaction rate of approx. 6000±200 M-1 s-1 in serum at 37 °C. To maximize the fluorescence signal, up to six trans-cyclooctene moieties were attached to the antibody used to pretarget cells for labeling. Com. available cetuximab was labeled with trans-cyclooctene succinimidyl carbonate and used for pretargeting expts.
- 18Agard, N. J.; Baskin, J. M.; Prescher, J. A.; Lo, A.; Bertozzi, C. R. A comparative study of bioorthogonal reactions with azides. ACS Chem. Biol. 2006, 1, 644– 648, DOI: 10.1021/cb6003228Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtFyhtbfJ&md5=fdee8107c9514d2cc3569d4d5b3df256A comparative study of bioorthogonal reactions with azidesAgard, Nicholas J.; Baskin, Jeremy M.; Prescher, Jennifer A.; Lo, Anderson; Bertozzi, Carolyn R.ACS Chemical Biology (2006), 1 (10), 644-648CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Detection of metabolites and post-translational modifications can be achieved using the azide as a bioorthogonal chem. reporter. Once introduced into target biomols., either metabolically or through chem. modification, the azide can be tagged with probes using one of three highly selective reactions: the Staudinger ligation, the Cu(I)-catalyzed azide-alkyne cycloaddn., or the strain-promoted [3+2] cycloaddn. Here, the authors compared these chemistries in the context of various biol. applications, including labeling of biomols. in complex lysates and on live cell surfaces. The Cu(I)-catalyzed reaction was most efficient for detecting azides in protein samples but was not compatible with live cells due to the toxicity of the reagents. Both the Staudinger ligation and the strain-promoted [3+2] cycloaddn. using optimized cyclooctynes were effective for tagging azides on live cells. The best reagent for this application was dependent upon the specific structure of the azide. These results provide a guide for biologists in choosing a suitable ligation chem.
- 19Dommerholt, J.; van Rooijen, O.; Borrmann, A.; Guerra, C. F.; Bickelhaupt, F. M.; van Delft, F. L. Highly accelerated inverse electron-demand cycloaddition of electron-deficient azides with aliphatic cyclooctynes. Nat. Commun. 2014, 5, 5378, DOI: 10.1038/ncomms6378Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvFansb7N&md5=d0038fe3424275c33a5ebfc561b2bce3Highly accelerated inverse electron-demand cycloaddition of electron-deficient azides with aliphatic cyclooctynesDommerholt, Jan; van Rooijen, Olivia; Borrmann, Annika; Guerra, Celia Fonseca; Bickelhaupt, F. Matthias; van Delft, Floris L.Nature Communications (2014), 5 (), 5378CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)Strain-promoted azide-alkyne cycloaddn. (SPAAC) as a conjugation tool has found broad application in material sciences, chem. biol. and even in vivo use. However, despite tremendous effort, SPAAC remains fairly slow (0.2-0.5 M-1 s-1) and efforts to increase reaction rates by tailoring of cyclooctyne structure have suffered from a poor trade-off between cyclooctyne reactivity and stability. We here wish to report tremendous acceleration of strain-promoted cycloaddn. of an aliph. cyclooctyne (bicyclo[6.1.0]non-4-yne, BCN) with electron-deficient aryl azides, with reaction rate consts. reaching 2.0-2.9 M-1 s-1. A remarkable difference in rate consts. of aliph. cyclooctynes vs. benzoannulated cyclooctynes is noted, enabling a next level of orthogonality by a judicious choice of azide-cyclooctyne combinations, which is inter alia applied in one-pot three-component protein labeling. The pivotal role of azide electronegativity is explained by d.-functional theory calcns. and electronic-structure analyses, which indicates an inverse electron-demand mechanism is operative with an aliph. cyclooctyne.
- 20Kamber, D. N.; Liang, Y.; Blizzard, R. J.; Liu, F.; Mehl, R. A.; Houk, K. N.; Prescher, J. A. 1,2,4-Triazines Are Versatile Bioorthogonal Reagents. J. Am. Chem. Soc. 2015, 137, 8388– 8391, DOI: 10.1021/jacs.5b05100Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtVansLnM&md5=43ead4d9e9086de4df148decd38e27121,2,4-Triazines Are Versatile Bioorthogonal ReagentsKamber, David N.; Liang, Yong; Blizzard, Robert J.; Liu, Fang; Mehl, Ryan A.; Houk, K. N.; Prescher, Jennifer A.Journal of the American Chemical Society (2015), 137 (26), 8388-8391CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)A new class of bioorthogonal reagents, 1,2,4-triazines, is described. These scaffolds are stable in biol. media and capable of robust reactivity with trans-cyclooctene (TCO). The enhanced stability of the triazine scaffold enabled its direct use in recombinant protein prodn. The triazine-TCO reaction can also be used in tandem with other bioorthogonal cycloaddn. reactions. These features fill current voids in the bioorthogonal toolkit.
- 21Hoffmann, J. E.; Plass, T.; Nikic, I.; Aramburu, I. V.; Koehler, C.; Gillandt, H.; Lemke, E. A.; Schultz, C. Highly Stable trans-Cyclooctene Amino Acids for Live-Cell Labeling. Chem. - Eur. J. 2015, 21, 12266– 12270, DOI: 10.1002/chem.201501647Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFOrtbjN&md5=632174adf34f4a3986213dadd02479fcHighly Stable trans-Cyclooctene Amino Acids for Live-Cell LabelingHoffmann, Jan-Erik; Plass, Tilman; Nikic, Ivana; Valle Aramburu, Iker; Koehler, Christine; Gillandt, Hartmut; Lemke, Edward A.; Schultz, CarstenChemistry - A European Journal (2015), 21 (35), 12266-12270CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)Trans-Cyclooctene groups incorporated into proteins via non-canonical amino acids (ncAAs) are emerging as specific handles for bioorthogonal chem. Here, we present a highly improved synthetic access to the axially and the equatorially linked trans-cyclooct-2-ene isomers (1 a,b). We further show that the axially connected isomer has a half-life about 10 times higher than the equatorial isomer and reacts with tetrazines much faster, as detd. by stopped-flow expts. The improved properties resulted in different labeling performance of the insulin receptor on the surface of intact cells.
- 22Rossin, R.; van den Bosch, S. M.; Ten Hoeve, W.; Carvelli, M.; Versteegen, R. M.; Lub, J.; Robillard, M. S. Highly reactive trans-cyclooctene tags with improved stability for Diels-Alder chemistry in living systems. Bioconjugate Chem. 2013, 24, 1210– 1217, DOI: 10.1021/bc400153yGoogle Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXosFygtrY%253D&md5=04e742d34442b4d0e8704a9be92d1e5fHighly Reactive trans-Cyclooctene Tags with Improved Stability for Diels-Alder Chemistry in Living SystemsRossin, Raffaella; van den Bosch, Sandra M.; ten Hoeve, Wolter; Carvelli, Marco; Versteegen, Ron M.; Lub, Johan; Robillard, Marc S.Bioconjugate Chemistry (2013), 24 (7), 1210-1217CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)One of the challenges of pretargeted radioimmunotherapy, which centers on the capture of a radiolabeled probe by a preinjected tumor-bound antibody, is the potential immunogenicity of biol. capturing systems. A bioorthogonal chem. approach may circumvent this drawback, but effective in vivo chem. in mice, larger animals, and eventually humans, requires very high reagent reactivity, sufficient stability, and retained selectivity. We report here that the reactivity of the fastest bioorthogonal reaction, the inverse-electron-demand-Diels-Alder cycloaddn. between a tetrazine probe and a trans-cyclooctene-tagged antibody, can be increased 10-fold (k2 = 2.7 × 105 M-1s-1) via the trans-cyclooctene, approaching the speed of biol. interactions, while also increasing its stability. This was enabled by the finding that the trans-cyclooctene tag is probably deactivated through isomerization to the unreactive cis-cyclooctene isomer by interactions with copper-contg. proteins, and that increasing the steric hindrance on the tag can impede this process. Next, we found that the higher reactivity of axial vs. equatorial linked TCO can be augmented by the choice of linker. The new, stabilized, and more reactive tag allowed for improved tumor-to-nontumor ratios in pretargeted tumor-bearing mice.
- 23Sletten, E. M.; Bertozzi, C. R. From mechanism to mouse: a tale of two bioorthogonal reactions. Acc. Chem. Res. 2011, 44, 666– 676, DOI: 10.1021/ar200148zGoogle Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtVWhurnF&md5=75f5134a99b940e43822840294bc9a5dFrom Mechanism to Mouse: A Tale of Two Bioorthogonal ReactionsSletten, Ellen M.; Bertozzi, Carolyn R.Accounts of Chemical Research (2011), 44 (9), 666-676CODEN: ACHRE4; ISSN:0001-4842. (American Chemical Society)A review. Bioorthogonal reactions are chem. reactions that neither interact with nor interfere with a biol. system. The participating functional groups must be inert to biol. moieties, must selectively reactive with each other under biocompatible conditions, and, for in vivo applications, must be nontoxic to cells and organisms. Addnl., it is helpful if one reactive group is small and therefore minimally perturbing of a biomol. into which it has been introduced either chem. or biosynthetically. Examples from the past decade suggest that a promising strategy for bioorthogonal reaction development begins with an anal. of functional group and reactivity space outside those defined by Nature. Issues such as stability of reactants and products (particularly in water), kinetics, and unwanted side reactivity with biofunctionalities must be addressed, ideally guided by detailed mechanistic studies. Finally, the reaction must be tested in a variety of environments, escalating from aq. media to biomol. solns. to cultured cells and, for the most optimized transformations, to live organisms. Work in our lab. led to the development of two bioorthogonal transformations that exploit the azide as a small, abiotic, and bioinert reaction partner: the Staudinger ligation and strain-promoted azide-alkyne cycloaddn. The Staudinger ligation is based on the classic Staudinger redn. of azides with triarylphosphines first reported in 1919. In the ligation reaction, the intermediate aza-ylide undergoes intramol. reaction with an ester, forming an amide bond faster than aza-ylide hydrolysis would otherwise occur in water. The Staudinger ligation is highly selective and reliably forms its product in environs as demanding as live mice. However, the Staudinger ligation has some liabilities, such as the propensity of phosphine reagents to undergo air oxidn. and the relatively slow kinetics of the reaction. The Staudinger ligation takes advantage of the electrophilicity of the azide; however, the azide can also participate in cycloaddn. reactions. In 1961, Wittig and Krebs noted that the strained, cyclic alkyne cyclooctyne reacts violently when combined neat with Ph azide, forming a triazole product by 1,3-dipolar cycloaddn. This observation stood in stark contrast to the slow kinetics assocd. with 1,3-dipolar cycloaddn. of azides with unstrained, linear alkynes, the conventional Huisgen process. Notably, the reaction of azides with terminal alkynes can be accelerated dramatically by copper catalysis (this highly popular Cu-catalyzed azide-alkyne cycloaddn. (CuAAC) is a quintessential "click" reaction). However, the copper catalysts are too cytotoxic for long-term exposure with live cells or organisms. Thus, for applications of bioorthogonal chem. in living systems, we built upon Wittig and Krebs' observation with the design of cyclooctyne reagents that react rapidly and selectively with biomol.-assocd. azides. This strain-promoted azide-alkyne cycloaddn. is often referred to as "Cu-free click chem.". Mechanistic and theor. studies inspired the design of a series of cyclooctyne compds. bearing fluorine substituents, fused rings, and judiciously situated heteroatoms, with the goals of optimizing azide cycloaddn. kinetics, stability, soly., and pharmacokinetic properties. Cyclooctyne reagents have now been used for labeling azide-modified biomols. on cultured cells and in live Caenorhabditis elegans, zebrafish, and mice. As this special issue testifies, the field of bioorthogonal chem. is firmly established as a challenging frontier of reaction methodol. and an important new instrument for biol. discovery. The above reactions, as well as several newcomers with bioorthogonal attributes, have enabled the high-precision chem. modification of biomols. in vitro, as well as real-time visualization of mols. and processes in cells and live organisms. The consequence is an impressive body of new knowledge and technol., amassed using a relatively small bioorthogonal reaction compendium. Expansion of this toolkit, an effort that is already well underway, is an important objective for chemists and biologists alike.
- 24Link, A. J.; Vink, M. K.; Tirrell, D. A. Presentation and detection of azide functionality in bacterial cell surface proteins. J. Am. Chem. Soc. 2004, 126, 10598– 10602, DOI: 10.1021/ja047629cGoogle Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXmtFOktL8%253D&md5=6bbc1f4e362bbe16dfd95b4620ef38abPresentation and detection of azide functionality in bacterial cell surface proteinsLink, A. James; Vink, Mandy K. S.; Tirrell, David A.Journal of the American Chemical Society (2004), 126 (34), 10598-10602CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)An improved protocol for copper-catalyzed triazole formation on the bacterial cell surface is described. Addn. of highly pure CuBr to cells treated with azidohomoalanine (2) leads to ∼. 10-Fold more extensive cell surface labeling than previously obsd. This highly active catalyst allows detection of the methionine analogs azidoalanine (1), azidonorvaline (3), and azidonorleucine (4) in cell surface proteins. Azidoalanine was previously believed to be silent with regard to the cellular protein synthesis machinery.
- 25Tian, F.; Tsao, M. L.; Schultz, P. G. A phage display system with unnatural amino acids. J. Am. Chem. Soc. 2004, 126, 15962– 15963, DOI: 10.1021/ja045673mGoogle Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXpslyksLk%253D&md5=32e9325687097a9bb9640da36ae8047cA Phage Display System with Unnatural Amino AcidsTian, Feng; Tsao, Meng-Lin; Schultz, Peter G.Journal of the American Chemical Society (2004), 126 (49), 15962-15963CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)We report a general method to display peptide-contg. unnatural amino acids on filamentous M13 phage. Five distinct unnatural amino acids were site-specifically incorporated at the N-terminal of the M13 phage minor coat protein pIII. Phages that contain p-azidophenylalanine can undergo a highly specific azide-alkyne [3+2] cycloaddn. reaction with an alkyne-derivatized fluorophore. The generalization of phage display to include unnatural amino acids should significantly increase the scope of phage display technol.
- 26Yang, J.; Seckute, J.; Cole, C. M.; Devaraj, N. K. Live-cell imaging of cyclopropene tags with fluorogenic tetrazine cycloadditions. Angew. Chem., Int. Ed. 2012, 51, 7476– 7479, DOI: 10.1002/anie.201202122Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xos1egs74%253D&md5=c60edddde8754df584b0205368be1e5cLive-Cell Imaging of Cyclopropene Tags with Fluorogenic Tetrazine CycloadditionsYang, Jun; Seckute, Jolita; Cole, Christian M.; Devaraj, Neal K.Angewandte Chemie, International Edition (2012), 51 (30), 7476-7479, S7476/1-S7476/26CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)The authors demonstrated the applicability of methylcyclopropene tags as dienophiles for reaction with fluorogenic tetrazines. Through systematic synthetic modifications, the stability, size, and reactivity of the cyclopropene scaffold were optimized. Methylcyclopropene derivs. were developed that react rapidly with tetrazines while retaining their aq. stability and small size. These cyclopropene handles elicited fluorescent responses from quenched tetrazine dyes and were suitable for cellular imaging applications, which was demonstrated by imaging cyclopropene phospholipids distributed in live human breast cancer cells.
- 27Patterson, D. M.; Nazarova, L. A.; Xie, B.; Kamber, D. N.; Prescher, J. A. Functionalized cyclopropenes as bioorthogonal chemical reporters. J. Am. Chem. Soc. 2012, 134, 18638– 18643, DOI: 10.1021/ja3060436Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsFWjurvL&md5=68598cbedefd059aaa5b63325d7d1ea6Functionalized Cyclopropenes as Bioorthogonal Chemical ReportersPatterson, David M.; Nazarova, Lidia A.; Xie, Bryan; Kamber, David N.; Prescher, Jennifer A.Journal of the American Chemical Society (2012), 134 (45), 18638-18643CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Chem. reporters are unique functional groups that can be used to label biomols. in living systems. Only a handful of broadly applicable reporters have been identified to date, owing to the rigorous demands placed on these functional groups in biol. settings. We describe here a new chem. reporter-cyclopropene-that can be used to target biomols. in vitro and in live cells. A variety of substituted cyclopropene scaffolds were synthesized and found to be stable in aq. soln. and in the presence of biol. nucleophiles. Furthermore, some of the cyclopropene units were metabolically introduced into cell surface glycans and subsequently detected with covalent probes. The small size and selective reactivity of cyclopropenes will facilitate efforts to tag diverse collections of biomols. in vivo.
- 28Andersen, K. A.; Aronoff, M. R.; McGrath, N. A.; Raines, R. T. Diazo groups endure metabolism and enable chemoselectivity in cellulo. J. Am. Chem. Soc. 2015, 137, 2412– 2415, DOI: 10.1021/ja5095815Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXitlWmsrs%253D&md5=de8c0a2a4bd272e46e60057cee01c3bdDiazo Groups Endure Metabolism and Enable Chemoselectivity in CelluloAndersen, Kristen A.; Aronoff, Matthew R.; McGrath, Nicholas A.; Raines, Ronald T.Journal of the American Chemical Society (2015), 137 (7), 2412-2415CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The authors introduce a stabilized diazo group as a reporter for chem. biol. ManDiaz, which is a diazo deriv. of N-acetylmannosamine, is found to endure cellular metab. and label the surface of a mammalian cell. There its diazo group can undergo a 1,3-dipolar cycloaddn. with a strained alkyne, providing a signal comparable to that from the azido congener, ManNAz. The chemoselectivity of diazo and alkynyl groups enables dual labeling of cells that is not possible with azido and alkynyl groups. Thus, the diazo group, which is approx. half the size of an azido group, provides unique opportunities for orthogonal labeling of cellular components.
- 29Karver, M. R.; Weissleder, R.; Hilderbrand, S. A. Bioorthogonal reaction pairs enable simultaneous, selective, multi-target imaging. Angew. Chem., Int. Ed. 2012, 51, 920– 922, DOI: 10.1002/anie.201104389Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs1SqurfM&md5=d8e69da77db3f495bc3422bd051dfd77Bioorthogonal Reaction Pairs Enable Simultaneous, Selective, Multi-Target ImagingKarver, Mark R.; Weissleder, Ralph; Hilderbrand, Scott A.Angewandte Chemie, International Edition (2012), 51 (4), 920-922, S920/1-S920/12CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)The authors describe the testing and in cell validation of a tetrazine-trans-cyclooctene (TCO) reaction pair that is orthogonal to azide-cyclooctyne cycloaddn. chem. The chosen pairs were able to react concurrently in the same culture to fluorescently label two different cancer cell types. The authors' method uses only small mol. based reagents that also can be readily incorporated into a wide variety of systems and potentially may be used in conjunction with fluorogenic azide and tetrazine substrates to achieve addnl. improvements in specificity and sensitivity. The ability to perform multiple, rapid, simultaneous chem. reactions with a high degree of specificity in chem. complex environments should prove to be a useful tool in chem., biol., and medicine.
- 30Cole, C. M.; Yang, J.; Seckute, J.; Devaraj, N. K. Fluorescent live-cell imaging of metabolically incorporated unnatural cyclopropene-mannosamine derivatives. ChemBioChem 2013, 14, 205– 208, DOI: 10.1002/cbic.201200719Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXjsleltg%253D%253D&md5=10f4ee328411eac99eab0797301994acFluorescent Live-Cell Imaging of Metabolically Incorporated Unnatural Cyclopropene-Mannosamine DerivativesCole, Christian M.; Yang, Jun; Seckute, Jolita; Devaraj, Neal K.ChemBioChem (2013), 14 (2), 205-208CODEN: CBCHFX; ISSN:1439-4227. (Wiley-VCH Verlag GmbH & Co. KGaA)This study reported that unnatural cyclo-propene-mannosamine derivs. can be used to image glycans on live human cancer cell lines. The results showed that recently developed methylcyclopropene bioorthogonal handles can be utilized for metabolic imaging of unnatural mannosamine derivs. on live-cell surfaces. In general, the data exhibit the possible use of cyclopropenes as reactive mini-tags for a myriad of applications in the profiling and imaging of small mols.
- 31Spate, A. K.; Busskamp, H.; Niederwieser, A.; Schart, V. F.; Marx, A.; Wittmann, V. Rapid labeling of metabolically engineered cell-surface glycoconjugates with a carbamate-linked cyclopropene reporter. Bioconjugate Chem. 2014, 25, 147– 154, DOI: 10.1021/bc4004487Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2c3lvVCqsg%253D%253D&md5=32429d08597db5715b49a065da7df2a1Rapid labeling of metabolically engineered cell-surface glycoconjugates with a carbamate-linked cyclopropene reporterSpate Anne-Katrin; Busskamp Holger; Niederwieser Andrea; Schart Verena F; Marx Andreas; Wittmann ValentinBioconjugate chemistry (2014), 25 (1), 147-54 ISSN:.Metabolic oligosaccharide engineering is a valuable tool to monitor cellular carbohydrates. Here, we report the synthesis of a novel N-acyl-mannosamine derivative bearing a methylcyclopropene tag that is attached to the sugar via a carbamate moiety. This derivative undergoes rapid Diels-Alder reaction with inverse electron demand. We demonstrate that the cell's biosynthetic machinery incorporates this non-natural mannosamine derivative into glycoconjugates that can, subsequently, be labeled within less than 10 min with a new sulfo-Cy3-tetrazine conjugate. Using this tetrazine-dye conjugate for the detection of the methylcyclopropene-tagged mannosamine derivative, we could achieve dual labeling of two different metabolically incorporated sugars combining a Diels-Alder reaction with inverse electron demand and a strain-promoted azide-alkyne cycloaddition which are carried out simultaneously in a single step.
- 32Sachdeva, A.; Wang, K.; Elliott, T.; Chin, J. W. Concerted, rapid, quantitative, and site-specific dual labeling of proteins. J. Am. Chem. Soc. 2014, 136, 7785– 7788, DOI: 10.1021/ja4129789Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXos1Wlurg%253D&md5=58e3a3c448deb78bf2a2e88ed57a8f9aConcerted, Rapid, Quantitative, and Site-Specific Dual Labeling of ProteinsSachdeva, Amit; Wang, Kaihang; Elliott, Thomas; Chin, Jason W.Journal of the American Chemical Society (2014), 136 (22), 7785-7788CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Rapid, one-pot, concerted, site-specific labeling of proteins at genetically encoded unnatural amino acids with distinct small mols. at physiol. pH, temp., and pressure is an important challenge. Current approaches require sequential labeling, low pH, and typically days to reach completion, limiting their utility. We report the efficient, genetically encoded incorporation of alkyne- and cyclopropene-contg. amino acids at distinct sites in a protein using an optimized orthogonal translation system in E. coli. and quant., site-specific, one-pot, concerted protein labeling with fluorophores bearing azide and tetrazine groups, resp. Protein double labeling in aq. buffer at physiol. pH, temp., and pressure is quant. in 30 min.
- 33Rideout, D. Self-assembling cytotoxins. Science 1986, 233, 561– 563, DOI: 10.1126/science.3523757Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL28Xlt12isbc%253D&md5=3abf3eb1b14176e35c5fd91193d5e3bdSelf-assembling cytotoxinsRideout, DarrylScience (Washington, DC, United States) (1986), 233 (4763), 561-3CODEN: SCIEAS; ISSN:0036-8075.Decanal [112-31-2] and N-amino-N'-1-octylguanidine (AOG) [66002-80-0], combined at 28 μM each, mediated erythrocyte lysis within 80 min under physiol. conditions. By contrast, no lysis was obsd. after 20 h with decanal (56 μM) or AOG (100 μM) alone. The pronounced synergism obsd. for these chems. and similar reactive pairs of chems. is due to the self-assembly of more cytotoxic hydrazones in situ. Decanal and AOG also exhibit synergistic activity against cultured human cells (HeLa) and bacteria (Escherichia coli J96). This synergism may be useful in the design of cytotoxins that would self-assemble selectively from nontoxic precursors within tumors, while sparing normal tissue.
- 34Murray, C. W.; Rees, D. C. The rise of fragment-based drug discovery. Nat. Chem. 2009, 1, 187– 192, DOI: 10.1038/nchem.217Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXmtF2rs78%253D&md5=0bd1a40fd927cce9200f0fa296a7462fThe rise of fragment-based drug discoveryMurray, Christopher W.; Rees, David C.Nature Chemistry (2009), 1 (3), 187-192CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)A review. The search for new drugs is plagued by high attrition rates at all stages in research and development. Chemists have an opportunity to tackle this problem because attrition can be traced back, in part, to the quality of the chem. leads. Fragment-based drug discovery (FBDD) is a new approach, increasingly used in the pharmaceutical industry, for reducing attrition and providing leads for previously intractable biol. targets. FBDD identifies low-mol.-wt. ligands (∼150 Da) that bind to biol. important macromols. The three-dimensional exptl. binding mode of these fragments is detd. using X-ray crystallog. or NMR spectroscopy, and is used to facilitate their optimization into potent mols. with drug-like properties. Compared with high-throughput-screening, the fragment approach requires fewer compds. to be screened, and, despite the lower initial potency of the screening hits, offers more efficient and fruitful optimization campaigns. Here, we review the rise of FBDD, including its application to discovering clin. candidates against targets for which other chem. approaches have struggled.
- 35Thirumurugan, P.; Matosiuk, D.; Jozwiak, K. Click chemistry for drug development and diverse chemical-biology applications. Chem. Rev. 2013, 113, 4905– 4979, DOI: 10.1021/cr200409fGoogle Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXkslahtLY%253D&md5=d9c664c199b63c7d556c60fe9cd1507dClick Chemistry for Drug Development and Diverse Chemical-Biology ApplicationsThirumurugan, Prakasam; Matosiuk, Dariusz; Jozwiak, KrzysztofChemical Reviews (Washington, DC, United States) (2013), 113 (7), 4905-4979CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review with 303 refs. on application of lick chem. reactions in chem., biochem., and pharmacol.
- 36Lewis, W. G.; Green, L. G.; Grynszpan, F.; Radic, Z.; Carlier, P. R.; Taylor, P.; Finn, M. G.; Sharpless, K. B. Click chemistry in situ: acetylcholinesterase as a reaction vessel for the selective assembly of a femtomolar inhibitor from an array of building blocks. Angew. Chem., Int. Ed. 2002, 41, 1053– 1057, DOI: 10.1002/1521-3773(20020315)41:6<1053::AID-ANIE1053>3.0.CO;2-4Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XisVeisbo%253D&md5=e1ff6aedcbf7ae1523f4e3082e801c86Click chemistry in situ: Acetylcholinesterase as a reaction vessel for the selective assembly of a femtomolar inhibitor from an array of building blocksLewis, Warren G.; Green, Luke G.; Grynszpan, Flavio; Radic, Zoran; Carlier, Paul R.; Taylor, Palmer; Finn, M. G.; Sharpless, K. BarryAngewandte Chemie, International Edition (2002), 41 (6), 1053-1057CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH)Form-fitting chem. in a protein mold is enabled by the use of the 1,3-dipolar cycloaddn. of azides and alkynes. The enzyme acetylcholinesterase preferentially assembles one pair of these reactants, each of which bears a group that binds to adjacent positions on the protein structure, into a 1,2,3-triazole adduct that is the most potent noncovalent inhibitor of the enzyme yet developed.
- 37Mocharla, V. P.; Colasson, B.; Lee, L. V.; Roper, S.; Sharpless, K. B.; Wong, C. H.; Kolb, H. C. In situ click chemistry: enzyme-generated inhibitors of carbonic anhydrase II. Angew. Chem., Int. Ed. 2005, 44, 116– 120, DOI: 10.1002/anie.200461580Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXptFaj&md5=9b890f3eecd0ede6132a45c90aff9607In situ click chemistry: Enzyme-generated inhibitors of carbonic anhydrase IIMocharla, Vani P.; Colasson, Benoit; Lee, Lac V.; Roeper, Stefanie; Sharpless, K. Barry; Wong, Chi-Huey; Kolb, Hartmuth C.Angewandte Chemie, International Edition (2005), 44 (1), 116-120CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Target-guided synthesis: Rather than making and screening thousands of compds. for lead discovery, in situ click chem. employs the biol. target itself to assemble its inhibitors by selectively binding and interconnecting reagents within the confines of its binding sites. Subnanomolar inhibitors of carbonic anhydrase II were produced by the enzyme from simple azide and acetylene precursors.
- 38Whiting, M.; Muldoon, J.; Lin, Y. C.; Silverman, S. M.; Lindstrom, W.; Olson, A. J.; Kolb, H. C.; Finn, M. G.; Sharpless, K. B.; Elder, J. H.; Fokin, V. V. Inhibitors of HIV-1 protease by using in situ click chemistry. Angew. Chem., Int. Ed. 2006, 45, 1435– 1439, DOI: 10.1002/anie.200502161Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XitF2qtLg%253D&md5=d575780546dbc314eb9df85bc2459539Inhibitors of HIV-1 protease by using in situ click chemistryWhiting, Matthew; Muldoon, John; Lin, Ying-Chuan; Silverman, Steven M.; Lindstrom, William; Olson, Arthur J.; Kolb, Hartmuth C.; Finn, M. G.; Sharpless, K. Barry; Elder, John H.; Fokin, Valery V.Angewandte Chemie, International Edition (2006), 45 (9), 1435-1439CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Twice poor equals potent: HIV-1 Protease assembles its own potent inhibitor through formation of the triazole linkage from azide- and alkyne-contg. fragments that are themselves poor binders.
- 39Devaraj, N. K.; Weissleder, R. Biomedical applications of tetrazine cycloadditions. Acc. Chem. Res. 2011, 44, 816– 827, DOI: 10.1021/ar200037tGoogle Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXmslOnu74%253D&md5=7c8ef732eea8dbf9e11b8bd4e4ddeef0Biomedical Applications of Tetrazine CycloadditionsDevaraj, Neal K.; Weissleder, RalphAccounts of Chemical Research (2011), 44 (9), 816-827CODEN: ACHRE4; ISSN:0001-4842. (American Chemical Society)A review. Disease mechanisms are increasingly being resolved at the mol. level. Biomedical success at this scale creates synthetic opportunities for combining specifically designed orthogonal reactions in applications such as imaging, diagnostics, and therapy. For practical reasons, it would be helpful if bioorthogonal coupling reactions proceeded with extremely rapid kinetics (k > 103 M-1 s-1) and high specificity. Improving kinetics would minimize both the time and amt. of labeling agent required to maintain high coupling yields. In this Account, we discuss our recent efforts to design extremely rapid bioorthogonal coupling reactions between tetrazines and strained alkenes. These selective reactions were first used to covalently couple conjugated tetrazine near-IR-emitting fluorophores to dienophile-modified extracellular proteins on living cancer cells. Confocal fluorescence microscopy demonstrated efficient and selective labeling, and control expts. showed minimal background fluorescence. Multistep techniques were optimized to work with nanomolar concns. of labeling agent over a time scale of minutes: the result was successful real-time imaging of covalent modification. We subsequently discovered fluorogenic probes that increase in fluorescence intensity after the chem. reaction, leading to an improved signal-to-background ratio. Fluorogenic probes were used for intracellular imaging of dienophiles. We further developed strategies to react and image chemotherapeutics, such as trans-cyclooctene taxol analogs, inside living cells. Because the coupling partners are small mols. (<300 Da), they offer unique steric advantages in multistep amplification. We also describe recent success in using tetrazine reactions to label biomarkers on cells with magneto-fluorescent nanoparticles. Two-step protocols that use bioorthogonal chem. can significantly amplify signals over both one-step labeling procedures as well as two-step procedures that use more sterically hindered biotin-avidin interactions. Nanoparticles can be detected with fluorescence or magnetic resonance techniques. These strategies are now being routinely used on clin. samples for biomarker profiling to predict malignancy and patient outcome. Finally, the authors discuss recent results with tetrazine reactions used for in vivo mol. imaging applications. Rapid tetrazine cycloaddns. allow modular labeling of small mols. with the most commonly used positron emission tomog. isotope, 18F. Addnl., recent work has applied this reaction directly in vivo for the pretargeted imaging of solid tumors. Future work with tetrazine cycloaddns. will undoubtedly lead to optimized protocols, improved probes, and addnl. biomedical applications.
- 40Lebraud, H.; Wright, D. J.; Johnson, C. N.; Heightman, T. D. Protein Degradation by In-Cell Self-Assembly of Proteolysis Targeting Chimeras. ACS Cent. Sci. 2016, 2, 927– 934, DOI: 10.1021/acscentsci.6b00280Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvFOltbbJ&md5=4cc9b4aaba6ee03cd2d4ea97a72728a5Protein Degradation by In-Cell Self-Assembly of Proteolysis Targeting ChimerasLebraud, Honorine; Wright, David J.; Johnson, Christopher N.; Heightman, Tom D.ACS Central Science (2016), 2 (12), 927-934CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)Selective degrdn. of proteins by proteolysis targeting chimeras (PROTACs) offers a promising potential alternative to protein inhibition for therapeutic intervention. Current PROTAC mols. incorporate a ligand for the target protein, a linker, and an E3 ubiquitin ligase recruiting group, which bring together target protein and ubiquitinating machinery. Such hetero-bifunctional mols. require significant linker optimization and possess high mol. wt., which can limit cellular permeation, soly., and other drug-like properties. We show here that the hetero-bifunctional mol. can be formed intracellularly by bio-orthogonal click combination of two smaller precursors. We designed a tetrazine tagged thalidomide deriv. which reacts rapidly with a trans-cyclo-octene tagged ligand of the target protein in cells to form a cereblon E3 ligase recruiting PROTAC mol. The in-cell click-formed proteolysis targeting chimeras (CLIPTACs) were successfully used to degrade two key oncol. targets, BRD4 and ERK1/2. ERK1/2 degrdn. was achieved using a CLIPTAC based on a covalent inhibitor. We expect this approach to be readily extendable to other inhibitor-protein systems because the tagged E3 ligase recruiter is capable of undergoing the click reaction with a suitably tagged ligand of any protein of interest to elicit its degrdn.
- 41Versteegen, R. M.; Rossin, R.; ten Hoeve, W.; Janssen, H. M.; Robillard, M. S. Click to release: instantaneous doxorubicin elimination upon tetrazine ligation. Angew. Chem., Int. Ed. 2013, 52, 14112– 14116, DOI: 10.1002/anie.201305969Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvVClsb%252FM&md5=c926aeb4b257768962ca1b3d6285a0aaClick to Release: Instantaneous Doxorubicin Elimination upon Tetrazine LigationVersteegen, Ron M.; Rossin, Raffaella; ten Hoeve, Wolter; Janssen, Henk M.; Robillard, Marc S.Angewandte Chemie, International Edition (2013), 52 (52), 14112-14116CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)We have developed a new bioorthogonal elimination reaction that enables instantaneous, self-immolative, and traceless release of doxorubicin from trans-cyclooctene following tetrazine ligation. In this proof-of-principle study, the inverse-electron-demand Diels-Alder (inv-DA) pyridazine elimination reached 79 % yield within minutes under ambient conditions at micromolar concns.
- 42Xu, M.; Tu, J.; Franzini, R. M. Rapid and efficient tetrazine-induced drug release from highly stable benzonorbornadiene derivatives. Chem. Commun. 2017, 53, 6271– 6274, DOI: 10.1039/C7CC03477FGoogle Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXosVymu7s%253D&md5=3b00491f3ba7b323ed3e1a6b84e8950aRapid and efficient tetrazine-induced drug release from highly stable benzonorbornadiene derivativesXu, Minghao; Tu, Julian; Franzini, Raphael M.Chemical Communications (Cambridge, United Kingdom) (2017), 53 (46), 6271-6274CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)A novel class of bioorthogonal release reactions based on benzonorbornadiene derivs. was developed. These carrier mols. are highly stable at physiol. conditions, but react rapidly with 1,2,4,5-tetrazines, and near-quant. release cargo mols. such as drugs and optical reporters.
- 43Neumann, K.; Jain, S.; Gambardella, A.; Walker, S. E.; Valero, E.; Lilienkampf, A.; Bradley, M. Tetrazine-Responsive Self-immolative Linkers. ChemBioChem 2017, 18, 91– 95, DOI: 10.1002/cbic.201600560Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitVyqsr7I&md5=d677f5477f4872930382cf2e1d3bb12eTetrazine-Responsive Self-immolative LinkersNeumann, Kevin; Jain, Sarthak; Gambardella, Alessia; Walker, Sarah E.; Valero, Elsa; Lilienkampf, Annamaria; Bradley, MarkChemBioChem (2017), 18 (1), 91-95CODEN: CBCHFX; ISSN:1439-4227. (Wiley-VCH Verlag GmbH & Co. KGaA)Mols. that undergo activation or modulation following the addn. of benign external small-mol. chem. stimuli have numerous applications. Here, we report the highly efficient "decaging" of a variety of moieties by activation of a "self-immolative" linker, by application of water-sol. and stable tetrazine, including the controlled delivery of doxorubicin in a cellular context.
- 44Jimenez-Moreno, E.; Guo, Z.; Oliveira, B. L.; Albuquerque, I. S.; Kitowski, A.; Guerreiro, A.; Boutureira, O.; Rodrigues, T.; Jimenez-Oses, G.; Bernardes, G. J. Vinyl Ether/Tetrazine Pair for the Traceless Release of Alcohols in Cells. Angew. Chem., Int. Ed. 2017, 56, 243– 247, DOI: 10.1002/anie.201609607Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitVGhtrzP&md5=e22f3ae8a0963e02028a8ee180bc7d09Vinyl Ether/Tetrazine Pair for the Traceless Release of Alcohols in CellsJimenez-Moreno, Ester; Guo, Zijian; Oliveira, Bruno L.; Albuquerque, Ines S.; Kitowski, Annabel; Guerreiro, Ana; Boutureira, Omar; Rodrigues, Tiago; Jimenez-Oses, Gonzalo; Bernardes, Goncalo J. L.Angewandte Chemie, International Edition (2017), 56 (1), 243-247CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)The cleavage of a protecting group from a protein or drug under bioorthogonal conditions enables accurate spatiotemporal control over protein or drug activity. Disclosed herein is that vinyl ethers serve as protecting groups for alc.-contg. mols. and as reagents for bioorthogonal bond-cleavage reactions. A vinyl ether moiety was installed in a range of mols., including amino acids, a monosaccharide, a fluorophore, and an analog of the cytotoxic drug duocarmycin. Tetrazine-mediated decaging proceeded under biocompatible conditions with good yields and reasonable kinetics. Importantly, the nontoxic, vinyl ether duocarmycin double prodrug was successfully decaged in live cells to reinstate cytotoxicity. This bioorthogonal reaction presents broad applicability and may be suitable for in vivo applications.
- 45Mejia Oneto, J. M.; Khan, I.; Seebald, L.; Royzen, M. In Vivo Bioorthogonal Chemistry Enables Local Hydrogel and Systemic Pro-Drug To Treat Soft Tissue Sarcoma. ACS Cent. Sci. 2016, 2, 476– 482, DOI: 10.1021/acscentsci.6b00150Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtFCqs77O&md5=b7a01ddbbf126ed981be5bb0dc2226a8In Vivo Bioorthogonal Chemistry Enables Local Hydrogel and Systemic Pro-Drug To Treat Soft Tissue SarcomaMejia Oneto, Jose M.; Khan, Irfan; Seebald, Leah; Royzen, MaksimACS Central Science (2016), 2 (7), 476-482CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)The ability to activate drugs only at desired locations avoiding systemic immunosuppression and other dose limiting toxicities is highly desirable. Here we present a new approach, named local drug activation, that uses bioorthogonal chem. to conc. and activate systemic small mols. at a location of choice. This method is independent of endogenous cellular or environmental markers and only depends on the presence of a preimplanted biomaterial near a desired site (e.g., tumor). We demonstrate the clear therapeutic benefit with minimal side effects of this approach in mice over systemic therapy using a doxorubicin pro-drug against xenograft tumors of a type of soft tissue sarcoma (HT1080).
- 46Zhang, G.; Li, J.; Xie, R.; Fan, X.; Liu, Y.; Zheng, S.; Ge, Y.; Chen, P. R. Bioorthogonal Chemical Activation of Kinases in Living Systems. ACS Cent. Sci. 2016, 2, 325– 331, DOI: 10.1021/acscentsci.6b00024Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmslWgsrw%253D&md5=ebb651cbb2fc3dca008863104c9a94d5Bioorthogonal Chemical Activation of Kinases in Living SystemsZhang, Gong; Li, Jie; Xie, Ran; Fan, Xinyuan; Liu, Yanjun; Zheng, Siqi; Ge, Yun; Chen, Peng R.ACS Central Science (2016), 2 (5), 325-331CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)Selective manipulation of protein kinases under living conditions is highly desirable yet extremely challenging, particularly in a gain-of-function fashion. Here we employ our recently developed bioorthogonal cleavage reaction as a general strategy for intracellular activation of individual kinases. Site-specific incorporation of trans-cyclooctene-caged lysine in place of the conserved catalytic lysine, in conjunction with the cleavage partner dimethyl-tetrazine, allowed efficient lysine decaging with the kinase activity chem. rescued in living systems.
- 47Li, J.; Jia, S.; Chen, P. R. Diels-Alder reaction-triggered bioorthogonal protein decaging in living cells. Nat. Chem. Biol. 2014, 10, 1003– 1005, DOI: 10.1038/nchembio.1656Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvVGiurfM&md5=4723cef7d02aec13267b61d08c9517a6Diels-Alder reaction-triggered bioorthogonal protein decaging in living cellsLi, Jie; Jia, Shang; Chen, Peng R.Nature Chemical Biology (2014), 10 (12), 1003-1005CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Small mols. that specifically activate an intracellular protein of interest are highly desirable. A generally applicable strategy, however, remains elusive. Herein we describe a small mol.-triggered bioorthogonal protein decaging technique that relies on the inverse electron-demand Diels-Alder reaction for eliminating a chem. caged protein side chain within living cells. This method permits the efficient activation of a given protein (for example, an enzyme) in its native cellular context within minutes.
- 48Wu, H.; Alexander, S. C.; Jin, S.; Devaraj, N. K. A Bioorthogonal Near-Infrared Fluorogenic Probe for mRNA Detection. J. Am. Chem. Soc. 2016, 138, 11429– 11432, DOI: 10.1021/jacs.6b01625Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xhtlamt77K&md5=5d18f87589f5d989b9b00655dd8b0663A Bioorthogonal Near-Infrared Fluorogenic Probe for mRNA DetectionWu, Haoxing; Alexander, Seth C.; Jin, Shuaijiang; Devaraj, Neal K.Journal of the American Chemical Society (2016), 138 (36), 11429-11432CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)There is significant interest in developing methods that visualize and detect RNA in live cells. Bioorthogonal template driven tetrazine ligations could be a powerful route to visualizing nucleic acids in native cells, yet past work has been limited with respect to the diversity of fluorogens that can be activated via a tetrazine reaction. Herein we report a novel bioorthogonal tetrazine uncaging reaction that harnesses tetrazine reactivity to unmask vinyl ether caged fluorophores spanning the visible spectrum, including a near-IR (NIR) emitting cyanine dye. Vinyl ether caged fluorophores and tetrazine partners are conjugated to high affinity antisense nucleic acid probes, which show highly selective fluorogenic reactivity when annealed to their resp. target RNA sequences. A target sequence in the 3' untranslated region (3' UTR) of an expressed mRNA was successfully detected in live cells employing appropriate nucleic acid probes bearing a tetrazine reactive NIR fluorogen. Given the expansion of tetrazine fluorogenic chem. to NIR dyes, we believe highly selective proximity induced fluorogenic tetrazine reactions could find broad uses in illuminating endogenous biomols. in living cells.
- 49Ji, X.; Zhou, C.; Ji, K.; Aghoghovbia, R. E.; Pan, Z.; Chittavong, V.; Ke, B.; Wang, B. Click and Release: A Chemical Strategy toward Developing Gasotransmitter Prodrugs by Using an Intramolecular Diels-Alder Reaction. Angew. Chem., Int. Ed. 2016, 55, 15846– 15851, DOI: 10.1002/anie.201608732Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvFahtb3P&md5=a4888e92153a23269644879744e14a12Click and Release: A Chemical Strategy toward Developing Gasotransmitter Prodrugs by Using an Intramolecular Diels-Alder ReactionJi, Xingyue; Zhou, Cheng; Ji, Kaili; Aghoghovbia, Robert E.; Pan, Zhixiang; Chittavong, Vayou; Ke, Bowen; Wang, BingheAngewandte Chemie, International Edition (2016), 55 (51), 15846-15851CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Prodrug strategies have been proven to be a very effective way of addressing delivery problems. Much of the chem. in prodrug development relies on the ability to mask an appropriate functional group, which can be removed under appropriate conditions. However, developing org. prodrugs of gasotransmitters represent unique challenges. This is esp. true with carbon monoxide, which does not have an easy "handle" for bioreversible derivatization. By taking advantage of an intramol. Diels-Alder reaction, we have developed a prodrug strategy for prepns. of org. CO prodrugs (I, where X = O, NR1 and n = 1,2) that are stable during synthesis and storage, and yet readily release CO with tunable release rates under near physiol. conditions. The effectiveness of the CO prodrug system in delivering a sufficient quantity of CO for possible therapeutic applications has been studied using a cell culture anti-inflammatory assay and a colitis animal model. These studies fully demonstrate the proof of concept, and lay a strong foundation for further medicinal chem. work in developing org. CO prodrugs.
- 50Carlson, J. C. T.; Mikula, H.; Weissleder, R. Unraveling Tetrazine-Triggered Bioorthogonal Elimination Enables Chemical Tools for Ultrafast Release and Universal Cleavage. J. Am. Chem. Soc. 2018, 140, 3603– 3612, DOI: 10.1021/jacs.7b11217Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitVKrtr8%253D&md5=0330db3cb69894d1d11e2343c98a4da0Unraveling tetrazine-triggered bioorthogonal elimination enables chemical tools for ultrafast release and universal cleavageCarlson, Jonathan C. T.; Mikula, Hannes; Weissleder, RalphJournal of the American Chemical Society (2018), 140 (10), 3603-3612CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Recent developments in bond cleavage reactions have expanded the scope of bioorthogonal chem. beyond click ligation and enabled new strategies for probe activation and therapeutic delivery. These applications, however, remain in their infancy, with further innovations needed to achieve the efficiency required for versatile and broadly useful tools in vivo. Among these chemistries, the tetrazine/trans-cyclooctene click-to-release reaction has exemplary kinetics and adaptability but achieves only partial release and is incompletely understood, which has limited its application. Investigating the mechanistic features of this reaction's performance, we discovered profound pH sensitivity, exploited it with acid-functionalized tetrazines that both enhance and markedly accelerate release, and ultimately uncovered an unexpected dead-end isomer as the reason for poor release. Implementing facile methods to prevent formation of this dead end, we have achieved exceptional efficiency, with essentially complete release across the full scope of physiol. pH, potentiating drug-delivery strategies and expanding the dynamic range of bioorthogonal on/off control.
- 51El-Sagheer, A. H.; Brown, T. Synthesis and polymerase chain reaction amplification of DNA strands containing an unnatural triazole linkage. J. Am. Chem. Soc. 2009, 131, 3958– 3964, DOI: 10.1021/ja8065896Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXisVCjtL0%253D&md5=c64505e86e67b410762714ad9af48364Synthesis and Polymerase Chain Reaction Amplification of DNA Strands Containing an Unnatural Triazole LinkageEl-Sagheer, Afaf H.; Brown, TomJournal of the American Chemical Society (2009), 131 (11), 3958-3964CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)DNA strands contg. an unnatural T-triazole-T linkage have been synthesized by click DNA ligation between oligonucleotides with 3'-AZT and 5'-propargylamido dT and amplified efficiently by polymerase chain reaction (PCR) using several different polymerases. DNA sequencing of PCR amplicons and clones in two different sequence contexts revealed the presence of a single thymidine at the ligation site. The remarkable ability of thermostable polymerases to reproducibly copy DNA templates contg. such an unnatural backbone opens up intriguing possibilities in gene synthesis, genetic anal., biol., and nanotechnol.
- 52Budin, I.; Devaraj, N. K. Membrane assembly driven by a biomimetic coupling reaction. J. Am. Chem. Soc. 2012, 134, 751– 753, DOI: 10.1021/ja2076873Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs12hsL3I&md5=25dbc6361458e7ff63b6cec654587696Membrane Assembly Driven by a Biomimetic Coupling ReactionBudin, Itay; Devaraj, Neal K.Journal of the American Chemical Society (2012), 134 (2), 751-753CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)One of the major goals of synthetic biol. is the development of non-natural cellular systems. In this work, we describe a catalytic biomimetic coupling reaction capable of driving the de novo self-assembly of phospholipid membranes. Our system features a copper-catalyzed azide-alkyne cycloaddn. that results in the formation of a triazole-contg. phospholipid analog. Concomitant assembly of membranes occurs spontaneously, not requiring preexisting membranes to house catalysts or precursors. The substitution of efficient synthetic reactions for key biochem. processes may offer a general route toward synthetic biol. systems.
- 53Cendret, V.; Francois-Heude, M.; Mendez-Ardoy, A.; Moreau, V.; Fernandez, J. M.; Djedaini-Pilard, F. Design and synthesis of a ″click″ high-mannose oligosaccharide mimic emulating Man8 binding affinity towards Con A. Chem. Commun. 2012, 48, 3733– 3735, DOI: 10.1039/c2cc30773aGoogle ScholarThere is no corresponding record for this reference.
- 54Dawson, P. E.; Muir, T. W.; Clark-Lewis, I.; Kent, S. B. Synthesis of proteins by native chemical ligation. Science 1994, 266, 776– 779, DOI: 10.1126/science.7973629Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXitVGgtrw%253D&md5=1d352d817ab48851500610dcd32d1136Synthesis of proteins by native chemical ligationDawson, Philip E.; Muir, Tom W.; Clark-Lewis, Ian; Kent, Stephen B. H.Science (Washington, D. C.) (1994), 266 (5186), 776-9CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)A simple technique has been devised that allows the direct synthesis of native backbone proteins of moderate size. Chemoselective reaction of two unprotected peptide segments gives an initial thioester-linked species. Spontaneous rearrangement of this transient intermediate yields a full-length product with a native peptide bond at the ligation site. The utility of native chem. ligation was demonstrated by the one-step prepn. of a cytokine contg. multiple disulfides. The polypeptide ligation product was folded and oxidized to form the native disulfide-contg. protein mol. Native chem. ligation is an important step toward the general application of chem. to proteins.
- 55de Figueiredo, R. M.; Suppo, J. S.; Campagne, J. M. Nonclassical Routes for Amide Bond Formation. Chem. Rev. 2016, 116, 12029– 12122, DOI: 10.1021/acs.chemrev.6b00237Google Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2svktV2ktQ%253D%253D&md5=833714af5bdc3786efdb953edfae36ecNonclassical Routes for Amide Bond Formationde Figueiredo Renata Marcia; Suppo Jean-Simon; Campagne Jean-MarcChemical reviews (2016), 116 (19), 12029-12122 ISSN:.The present review offers an overview of nonclassical (e.g., with no pre- or in situ activation of a carboxylic acid partner) approaches for the construction of amide bonds. The review aims to comprehensively discuss relevant work, which was mainly done in the field in the last 20 years. Organization of the data follows a subdivision according to substrate classes: catalytic direct formation of amides from carboxylic and amines ( section 2 ); the use of carboxylic acid surrogates ( section 3 ); and the use of amine surrogates ( section 4 ). The ligation strategies (NCL, Staudinger, KAHA, KATs, etc.) that could involve both carboxylic acid and amine surrogates are treated separately in section 5 .
- 56Nilsson, B. L.; Kiessling, L. L.; Raines, R. T. High-yielding Staudinger ligation of a phosphinothioester and azide to form a peptide. Org. Lett. 2001, 3, 9– 12, DOI: 10.1021/ol006739vGoogle Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXovVOjurg%253D&md5=fff7cb4d30c89b152983e3e5f50f157dHigh-Yielding Staudinger Ligation of a Phosphinothioester and Azide To Form a PeptideNilsson, Bradley L.; Kiessling, Laura L.; Raines, Ronald T.Organic Letters (2001), 3 (1), 9-12CODEN: ORLEF7; ISSN:1523-7060. (American Chemical Society)The Staudinger ligation can be used to couple a peptide with a C-terminal phosphinothioester to another with an N-terminal α-azido group to form a single peptide that contains no residual atoms of the phosphinothiol entity. Here, diphenylphosphinomethanethiol thioesters are shown to give high isolated yields for this transformation. This finding provides precedent for a powerful and versatile new method for the total synthesis of proteins, avoiding the native chem. ligation method's reliance on the presence of a cysteine residue for bond formation.
- 57Saxon, E.; Armstrong, J. I.; Bertozzi, C. R. A ″traceless″ Staudinger ligation for the chemoselective synthesis of amide bonds. Org. Lett. 2000, 2, 2141– 2143, DOI: 10.1021/ol006054vGoogle Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXktVGktrk%253D&md5=88b7c588c348dd4b59b996db08c3d0a1A "Traceless" Staudinger Ligation for the Chemoselective Synthesis of Amide BondsSaxon, Eliana; Armstrong, Joshua I.; Bertozzi, Carolyn R.Organic Letters (2000), 2 (14), 2141-2143CODEN: ORLEF7; ISSN:1523-7060. (American Chemical Society)A novel modification is reported of the authors' previously reported Staudinger ligation that generates an amide bond from an azide and a specifically functionalized phosphine. This method for the selective formation of an amide bond, which does not require the orthogonal protection of distal functional groups, should find general utility in synthetic and biol. chem. 2-AcOC6H4PPh2 and (1-acetyl-2-imidazolyl)diphenylphosphine reacted with an azidonucleoside (5'-azido-N-benzoyl-2',5'-dideoxyadenosine) in wet THF to convert the azido group to an acetylamino group in >95% yield.
- 58Erben, A.; Grossmann, T. N.; Seitz, O. DNA-triggered synthesis and bioactivity of proapoptotic peptides. Angew. Chem., Int. Ed. 2011, 50, 2828– 2832, DOI: 10.1002/anie.201007103Google Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXivVGhurs%253D&md5=b4096818c6a8bd987fd1c8e7d913c661DNA-Triggered Synthesis and Bioactivity of Proapoptotic PeptidesErben, Anne; Grossmann, Tom N.; Seitz, OliverAngewandte Chemie, International Edition (2011), 50 (12), 2828-2832, S2828/1-S2828/17CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Diseases are frequently caused by changes in the genetic infrastructure. In such cases, the disordered state of a diseased cell is encoded in the DNA and reflected in the level and sequence of the expressed RNA mols. The information obtained from nucleic acids may be used to direct mol. therapies only to diseased cells and tissues. In a fascinating approach, disease-specific nucleic acid sequences could be hijacked to trigger the formation or release of drug mols. Herein a reaction system is introduced in which the sequence information of an unstructured DNA template is used to trigger the transfer of an aminoacyl group from a donating thioester-modified peptide-nucleic acid (PNA) conjugate to an acceptor peptidyl-PNA conjugate. It is demonstrated that the template can act as a catalyst which instructs the formation of many product mols. per template mol. The formed peptide-PNA conjugate was designed to interfere with the protein-protein interactions between caspase-9, a protease involved in the initiation of programmed cell death (apoptosis), and the X-linked inhibitor of apoptosis protein XIAP. It is shown that the nucleic acid programmed peptide synthesis allows activation of caspase-9 and a downstream caspase.
- 59Noda, H.; Eros, G.; Bode, J. W. Rapid ligations with equimolar reactants in water with the potassium acyltrifluoroborate (KAT) amide formation. J. Am. Chem. Soc. 2014, 136, 5611– 5614, DOI: 10.1021/ja5018442Google Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXltFGltLc%253D&md5=755cfa1e934bab498e8ac3c9d67d8009Rapid ligations with equimolar reactants in water with the potassium acyltrifluoroborate (KAT) amide formationNoda, Hidetoshi; Eros, Gabor; Bode, Jeffrey W.Journal of the American Chemical Society (2014), 136 (15), 5611-5614CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The identification of fast, chemoselective bond-forming reactions is one of the major contemporary challenges in chem. We show that chemoselective amide-forming ligations of potassium acyltrifluoroborates (KATs) and O-carbamoylhydroxylamines proceed in the presence of all unprotected functional groups with a second-order rate const. of 20 M-1 s-1. PEG chains, lipids, biotin, and dyes were introduced onto an unprotected 31-mer peptide (GLP-1 analog) with equimolar ratios of reactants within minutes at 1 mM and within 1 h at 100 μM, even with Mw 20 000 PEG. This conjugation reaction provides a new approach to the synthesis of mols. such as protein-protein and protein-polymer conjugates.
- 60Rudd, A. K.; Devaraj, N. K. Traceless Synthesis of Ceramides in Living Cells Reveals Saturation-Dependent Apoptotic Effects. Proc. Natl. Acad. Sci. USA , 2018, In Press. DOI: DOI: 10.1073/pnas.1804266115 .Google ScholarThere is no corresponding record for this reference.
- 61Mehl, R. A.; Anderson, J. C.; Santoro, S. W.; Wang, L.; Martin, A. B.; King, D. S.; Horn, D. M.; Schultz, P. G. Generation of a bacterium with a 21 amino acid genetic code. J. Am. Chem. Soc. 2003, 125, 935– 939, DOI: 10.1021/ja0284153Google Scholar61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXhtlCmug%253D%253D&md5=fdbba595b21142ebd43fbdaf399008aaGeneration of a Bacterium with a 21 Amino Acid Genetic CodeMehl, Ryan A.; Anderson, J. Christopher; Santoro, Stephen W.; Wang, Lei; Martin, Andrew B.; King, David S.; Horn, David M.; Schultz, Peter G.Journal of the American Chemical Society (2003), 125 (4), 935-939CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)We have generated a completely autonomous bacterium with a 21 amino acid genetic code. This bacterium can biosynthesize a nonstandard amino acid from basic carbon sources and incorporate this amino acid into proteins in response to the amber nonsense codon. The biosynthetic pathway for the amino acid p-aminophenylalanine (pAF) as well as a unique pAF synthetase and cognate tRNA were added to Escherichia coli. Denaturing gel electrophoresis and mass spectrometric anal. show that pAF is incorporated into myoglobin with fidelity and efficiency rivaling those of the common 20 amino acids. This and other such organisms may provide an opportunity to examine the evolutionary consequences of adding new amino acids to the genetic repertoire, as well as generate proteins with new or enhanced biol. functions.
- 62Bohannon, M. B.; Kleiman, R. Cyclopropene fatty acids of selected seed oils from bombacaceae, malvaceae, and sterculiaceae. Lipids 1978, 13, 270– 273, DOI: 10.1007/BF02533669Google ScholarThere is no corresponding record for this reference.
- 63Bao, X.; Katz, S.; Pollard, M.; Ohlrogge, J. Carbocyclic fatty acids in plants: biochemical and molecular genetic characterization of cyclopropane fatty acid synthesis of Sterculiafoetida. Proc. Natl. Acad. Sci. U. S. A. 2002, 99, 7172– 7177, DOI: 10.1073/pnas.092152999Google ScholarThere is no corresponding record for this reference.
- 64Chen, K.; Huang, X.; Kan, S. B. J.; Zhang, R. K.; Arnold, F. H. Enzymatic construction of highly strained carbocycles. Science 2018, 360, 71– 75, DOI: 10.1126/science.aar4239Google Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXmvFamsL0%253D&md5=df5bbb212f61f1f29ed867125d53dcdeEnzymatic construction of highly strained carbocyclesChen, Kai; Huang, Xiongyi; Kan, S. B. Jennifer; Zhang, Ruijie K.; Arnold, Frances H.Science (Washington, DC, United States) (2018), 360 (6384), 71-75CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Small carbocycles are structurally rigid and possess high intrinsic energy due to their ring strain. These features lead to broad applications but also create challenges for their construction. We report the engineering of a cytochrome P 450 variant (designated P411) that catalyze the formation of chiral bicyclobutanes, one of the most strained four-membered systems, via successive carbene addn. to unsatd. carbon-carbon bonds. Enzymes that produce cyclopropenes, putative intermediates to the bicyclobutanes, were also identified. These genetically encoded proteins are readily optimized by directed evolution, function in Escherichia coli, and act on structurally diverse substrates with high efficiency and selectivity, providing an effective route to many chiral strained structures. This biotransformation is easily performed at preparative scale, and the resulting strained carbocycles can be derivatized, opening myriad potential applications.
- 65Lappchen, T.; Rossin, R.; van Mourik, T. R.; Gruntz, G.; Hoeben, F. J. M.; Versteegen, R. M.; Janssen, H. M.; Lub, J.; Robillard, M. S. DOTA-tetrazine probes with modified linkers for tumor pretargeting. Nucl. Med. Biol. 2017, 55, 19– 26, DOI: 10.1016/j.nucmedbio.2017.09.001Google Scholar65https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1M7gtlCnug%253D%253D&md5=c8e55fde4872a6407e12fce3812fe901DOTA-tetrazine probes with modified linkers for tumor pretargetingLappchen Tilman; Rossin Raffaella; van Mourik Tiemen R; Gruntz Guillaume; Lub Johan; Hoeben Freek J M; Versteegen Ron M; Janssen Henk M; Robillard Marc SNuclear medicine and biology (2017), 55 (), 19-26 ISSN:.INTRODUCTION: Pretargeted radioimmunoimaging and -therapy approaches building on the bioorthogonal inverse-electron-demand Diels-Alder (IEDDA) reaction between strained trans-cyclooctenes (TCO) and electron-deficient tetrazines (Tz) have yielded impressive results in recent years and have proven a vital alternative to biological pretargeting systems. After improvement of the TCO-antibody conjugates, we here report on our evaluation of a new series of radiolabeled Tz-probes. METHODS: Four new Tz-probes were synthesized, radiolabeled with lutetium-177, and characterized in vitro in terms of lipophilicity, reactivity, and stability in PBS and mouse serum. The in vivo biodistribution profile and tumor-targeting potential of the probes were evaluated in LS174T tumor-bearing mice pretargeted with TCO-antibody conjugates using non-pretargeted mice as control. RESULTS: Radiolabeling of all probes proceeded in high yields providing the (177)Lu-labeled tetrazines in >95% radiochemical purity without any further purification. In mouse serum, half-lives of the probes varied between 8 and 13 h, with the exception of the most lipophilic probe, [(177)Lu]1b, with a serum half-life of less than 1 h. This probe also showed the fastest blood clearance (t1/2 = 5.4 min), more than 2-fold faster than PEG-linked probes [(177)Lu]3 and [(177)Lu]4, and even 3-fold faster than the other small probes without the PEG-linker, [(177)Lu]1a and [(177)Lu]2. In the pretargeting experiments, tumor uptake of the lead probe [(177)Lu]4 (~6 %ID/g) was most closely approached by [(177)Lu]2, followed by [(177)Lu]3 and [(177)Lu]1a. While all the smaller and more lipophilic probes suffered from increased liver uptake, the PEG-linked probe [(177)Lu]3 with its additional negative charge surprisingly showed the highest kidney uptake among all of the probes. CONCLUSION: The in vitro performance of some of the new tetrazine probes turned out to be comparable to the established lead probe [(177)Lu]Lu-DOTA-PEG11-Tz ([(177)Lu]4). However, tumor pretargeting studies in vivo showed lower tumor uptake and increased uptake in non-target organs.
- 66Houghton, J. L.; Membreno, R.; Abdel-Atti, D.; Cunanan, K. M.; Carlin, S.; Scholz, W. W.; Zanzonico, P. B.; Lewis, J. S.; Zeglis, B. M. Establishment of the In Vivo Efficacy of Pretargeted Radioimmunotherapy Utilizing Inverse Electron Demand Diels-Alder Click Chemistry. Mol. Cancer Ther. 2017, 16, 124– 133, DOI: 10.1158/1535-7163.MCT-16-0503Google Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjsF2hsQ%253D%253D&md5=eb72c9c0f7b38994358445c8586f9570Establishment of the In Vivo Efficacy of Pretargeted Radioimmunotherapy Utilizing Inverse Electron Demand Diels-Alder Click ChemistryHoughton, Jacob L.; Membreno, Rosemery; Abdel-Atti, Dalya; Cunanan, Kristen M.; Carlin, Sean; Scholz, Wolfgang W.; Zanzonico, Pat B.; Lewis, Jason S.; Zeglis, Brian M.Molecular Cancer Therapeutics (2017), 16 (1), 124-133CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)The pretargeting system based on the inverse electron demand Diels-Alder reaction (IEDDA) between trans-cyclooctene (TCO) and tetrazine (Tz) combines the favorable pharmacokinetic properties of radiolabeled small mols. with the affinity and specificity of antibodies. This strategy has proven to be an efficient method for the molecularly targeted delivery of pharmaceuticals, including isotopes for radiol. imaging. Despite encouraging results from in vivo PET imaging studies, this promising system has yet to be thoroughly evaluated for pretargeted radioimmunotherapy (PRIT). Toward that end, we synthesized two novel 177Lu-labeled tetrazine-bearing radioligands. Next, we compared the usefulness of our ligands for PRIT when paired with TCO-modified 5B1-a human, anti-CA19.9 mAb-in preclin. murine models of pancreatic cancer. The exemplary ligand, 177Lu-DOTA-PEG7-Tz, showed rapid (4.6 ± 0.8% ID/g at 4 h) and persistent (16.8 ± 3.9% ID/g at 120 h) uptake in tumors while concurrently clearing from blood and nontarget tissues. Single-dose therapy studies using 5B1-TCO and varying amts. of 177Lu-DOTA-PEG7-Tz (400, 800, and 1,200μCi) showed that our system elicits a dose-dependent therapeutic response in mice bearing human xenografts. Furthermore, dosimetry calcns. suggest that our approach is amenable to clin. applications with its excellent dosimetric profile in organs of clearance (i.e., liver and kidneys) as well as in dose-limiting tissues, such as red marrow. This study established that a pretargeted methodol. utilizing the IEDDA reaction can rapidly and specifically deliver a radiotherapeutic payload to tumor tissue, thus illustrating its excellent potential for clin. translation.
- 67Goodwin, D. A.; Meares, C. F.; McCall, M. J.; McTigue, M.; Chaovapong, W. Pre-targeted immunoscintigraphy of murine tumors with indium-111-labeled bifunctional haptens. J. Nucl. Med. 1988, 29 (2), 226– 234Google ScholarThere is no corresponding record for this reference.
- 68Zeglis, B. M.; Sevak, K. K.; Reiner, T.; Mohindra, P.; Carlin, S. D.; Zanzonico, P.; Weissleder, R.; Lewis, J. S. A pretargeted PET imaging strategy based on bioorthogonal Diels-Alder click chemistry. J. Nucl. Med. 2013, 54, 1389– 1396, DOI: 10.2967/jnumed.112.115840Google Scholar68https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVaqu7fF&md5=a930e7e2f9b22e030b692399ff53baafA pretargeted PET imaging strategy based on bioorthogonal Diels-Alder click chemistryZeglis, Brian M.; Sevak, Kuntal K.; Reiner, Thomas; Mohindra, Priya; Carlin, Sean D.; Zanzonico, Pat; Weissleder, Ralph; Lewis, Jason S.Journal of Nuclear Medicine (2013), 54 (8), 1389-1396CODEN: JNMEAQ; ISSN:0161-5505. (Society of Nuclear Medicine and Molecular Imaging)The specificity of antibodies have made immunoconjugates promising vectors for the delivery of radioisotopes to cancer cells; however, their long pharmacol. half-lives necessitate the use of radioisotopes with long phys. half-lives, a combination that leads to high radiation doses to patients. Therefore, the development of targeting modalities that harness the advantages of antibodies without their pharmacokinetic limitations is desirable. To this end, we report the development of a methodol. for pretargeted PET imaging based on the bioorthogonal Diels-Alder click reaction between tetrazine and transcyclooctene. Methods: A proof-of-concept system based on the A33 antibody, SW1222 colorectal cancer cells, and 64Cu was used. The huA33 antibody was covalently modified with transcyclooctene, and a NOTA-modified tetrazine was synthesized and radiolabeled with 64Cu. Pretargeted in vivo biodistribution and PET imaging expts. were performed with athymic nude mice bearing A33 antigen-expressing, SW1222 colorectal cancer xenografts. Results: The huA33 antibody was modified with transcyclooctene to produce a conjugate with high immunoreactivity, and the 64Cu-NOTA-labeled tetrazine ligand was synthesized with greater than 99% purity and a specific activity of 9-10 MBq/μg. For in vivo expts., mice bearing SW1222 xenografts were injected with transcyclooctene-modified A33; after allowing 24 h for accumulation of the antibody in the tumor, the mice were injected with 64Cu-NOTA-labeled tetrazine for PET imaging and biodistribution expts. At 12 h after injection, the retention of uptake in the tumor (4.1 ± 0.3 percent injected dose per g), coupled with the fecal excretion of excess radioligand, produced images with high tumor-to-background ratios. PET imaging and biodistribution expts. performed using A33 directly labeled with either 64Cu or 89Zr revealed that although abs. tumor uptake was higher with the directly radiolabeled antibodies, the pretargeted system yielded comparable images and tumor-to-muscle ratios at 12 and 24 h after injection. Further, dosimetry calcns. revealed that the 64Cu pretargeting system resulted in only a fraction of the absorbed background dose of A33 directly labeled with 89Zr (0.0124 mSv/MBq vs. 0.4162 mSv/MBq, resp.). Conclusion: The high quality of the images produced by this pretargeting approach, combined with the ability of the methodol. to dramatically reduce nontarget radiation doses to patients, marks this system as a strong candidate for clin. translation.
- 69Rossin, R.; Verkerk, P. R.; van den Bosch, S. M.; Vulders, R. C.; Verel, I.; Lub, J.; Robillard, M. S. In vivo chemistry for pretargeted tumor imaging in live mice. Angew. Chem., Int. Ed. 2010, 49, 3375– 3378, DOI: 10.1002/anie.200906294Google Scholar69https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXlt1Glsbo%253D&md5=5b08f7ad878d9f823dbdd6b4096b71e1In Vivo Chemistry for Pretargeted Tumor Imaging in Live MiceRossin, Raffaella; Renart Verkerk, Pascal; van den Bosch, Sandra M.; Vulders, Roland C. M.; Verel, Iris; Lub, Johan; Robillard, Marc S.Angewandte Chemie, International Edition (2010), 49 (19), 3375-3378, S3375/1-S3375/22CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Bioorthogonal chem. reaction between two exogenous moieties in living animal was used for cancer imaging.
- 70Devaraj, N. K.; Weissleder, R.; Hilderbrand, S. A. Tetrazine-based cycloadditions: application to pretargeted live cell imaging. Bioconjugate Chem. 2008, 19, 2297– 2299, DOI: 10.1021/bc8004446Google Scholar70https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsVWrtbfJ&md5=f68878b74748e037b97e9a1f37178250Tetrazine-Based Cycloadditions: Application to Pretargeted Live Cell ImagingDevaraj, Neal K.; Weissleder, Ralph; Hilderbrand, Scott A.Bioconjugate Chemistry (2008), 19 (12), 2297-2299CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Bioorthogonal tetrazine cycloaddns. have been applied to live cell labeling. Tetrazines react irreversibly with the strained dienophile norbornene forming dihydropyrazine products and dinitrogen. The reaction is high yielding, selective, and fast in aq. media. Her2/neu receptors on live human breast cancer cells were targeted with a monoclonal antibody modified with a norbornene. Tetrazines conjugated to a near-IR fluorochrome selectively and rapidly label the pretargeted antibody in the presence of serum. These findings indicate that this chem. is suitable for in vitro labeling expts., and suggests that it may prove a useful strategy for in vivo pretargeted imaging under numerous modalities.
Cited By
This article is cited by 390 publications.
- Mengxue Zheng, Wenxiu Gao, Lei Zhang, Kaiyu Yan, Zhenguo Zhang, Peizhong Xie, Teck-Peng Loh. Allylation of Lactol in Water. Organic Letters 2024, 26
(47)
, 10201-10206. https://doi.org/10.1021/acs.orglett.4c04189
- Krishna Vippala, Shreyas Shankar Wagle, Parul Rathee, Keerthana Mulamukkil, Yousif Ayoub, Arthur Komlosh, Sharon Gazal, Bianca Avramovitch, Roey J. Amir. Micellar “Click” Nanoreactors: Spiking Pluronic-Based Micelles with Polymeric Ligands. Macromolecules 2024, 57
(22)
, 10557-10566. https://doi.org/10.1021/acs.macromol.4c01425
- Ranjan Sasmal, Arka Som, Pratibha Kumari, Resmi V. Nair, Sushanta Show, Nisha Sanjay Barge, Meenakshi Pahwa, Nilanjana Das Saha, Sushma Rao, Sheeba Vasu, Rachit Agarwal, Sarit S. Agasti. Supramolecular Guest Exchange in Cucurbit[7]uril for Bioorthogonal Fluorogenic Imaging across the Visible Spectrum. ACS Central Science 2024, 10
(10)
, 1945-1959. https://doi.org/10.1021/acscentsci.4c01080
- Veronika Šlachtová, Simona Bellová, Milan Vrabel. Synthesis of C3-Substituted N1-tert-Butyl 1,2,4-Triazinium Salts via the Liebeskind–Srogl Reaction for Fluorogenic Labeling of Live Cells. The Journal of Organic Chemistry 2024, 89
(20)
, 14634-14640. https://doi.org/10.1021/acs.joc.3c02454
- Cheng Yang, Kai Lu, Jie Li, Haoxing Wu, Wei Chen. Rapid Construction of 18F-Triazolyl-tetrazines through the Click Reaction. The Journal of Organic Chemistry 2024, 89
(20)
, 14673-14678. https://doi.org/10.1021/acs.joc.4c00574
- Balasaheb R. Borade, Ming Fang, Qing Lin. Synthesis of Hydrazonyl Sultones via Phosphine-Mediated Cyclodehydration of Vicinal Sulfo-acyl Hydrazides. Organic Letters 2024, 26
(39)
, 8239-8243. https://doi.org/10.1021/acs.orglett.4c02687
- Haojie Yang, Hongbao Sun, Yinghan Chen, Yayue Wang, Cheng Yang, Fang Yuan, Xiaoai Wu, Wei Chen, Ping Yin, Yong Liang, Haoxing Wu. Enabling Universal Access to Rapid and Stable Tetrazine Bioorthogonal Probes through Triazolyl-Tetrazine Formation. JACS Au 2024, 4
(8)
, 2853-2861. https://doi.org/10.1021/jacsau.3c00843
- Soraya Learte-Aymamí, Laura Martínez-Castro, Carmen González-González, Miriam Condeminas, Pau Martin-Malpartida, María Tomás-Gamasa, Sandra Baúlde, José R. Couceiro, Jean-Didier Maréchal, Maria J. Macias, José L. Mascareñas, M. Eugenio Vázquez. De Novo Engineering of Pd-Metalloproteins and Their Use as Intracellular Catalysts. JACS Au 2024, 4
(7)
, 2630-2639. https://doi.org/10.1021/jacsau.4c00379
- Mohsen Khodadadi Yazdi, Farzad Seidi, Aleksander Hejna, Payam Zarrintaj, Navid Rabiee, Justyna Kucinska-Lipka, Mohammad Reza Saeb, Sidi A. Bencherif. Tailor-Made Polysaccharides for Biomedical Applications. ACS Applied Bio Materials 2024, 7
(7)
, 4193-4230. https://doi.org/10.1021/acsabm.3c01199
- Nathan Noël, Agathe Martinez, Fabien Massicot, Jean-Luc Vasse, Jean-Bernard Behr. Kinetics of Strain-Promoted Alkyne-Nitrone Cycloadditions (SPANC) with Unprotected Carbohydrate Scaffolded Nitrones. Organic Letters 2024, 26
(18)
, 3917-3922. https://doi.org/10.1021/acs.orglett.4c01098
- Murat Işık, Mehmet Ali Kısaçam. Readily Accessible and Brightly Fluorogenic BODIPY/NBD–Tetrazines via SNAr Reactions. The Journal of Organic Chemistry 2024, 89
(9)
, 6513-6519. https://doi.org/10.1021/acs.joc.3c02864
- Aysun Degirmenci, Rana Sanyal, Amitav Sanyal. Metal-Free Click-Chemistry: A Powerful Tool for Fabricating Hydrogels for Biomedical Applications. Bioconjugate Chemistry 2024, 35
(4)
, 433-452. https://doi.org/10.1021/acs.bioconjchem.4c00003
- Yun Hu, Rachel Spiegelhoff, Ken S. Lee, Kyana M. Sanders, Jennifer M. Schomaker. A Synthetic Strategy toward S-, N-, and O-Heterocyclooctynes Facilitates Bioconjugation Using Multifunctional Handles. The Journal of Organic Chemistry 2024, 89
(7)
, 4512-4522. https://doi.org/10.1021/acs.joc.3c02747
- Zhiguo Gao, Yaojia Li, Jiaqi Xing, Yougong Lu, Quanlin Shao, Jinzhong Hu, Shan Zhao, Wei He, Baiwang Sun. Transition Metal Ru(II) Catalysts Immobilized Nanoreactors for Conditional Bioorthogonal Catalysis in Cells. ACS Applied Materials & Interfaces 2024, 16
(13)
, 15870-15878. https://doi.org/10.1021/acsami.3c19133
- Amit Sharma, Peter Verwilst, Mingle Li, Dandan Ma, Nem Singh, Jiyoung Yoo, Yujin Kim, Ying Yang, Jing-Hui Zhu, Haiqiao Huang, Xi-Le Hu, Xiao-Peng He, Lintao Zeng, Tony D. James, Xiaojun Peng, Jonathan L. Sessler, Jong Seung Kim. Theranostic Fluorescent Probes. Chemical Reviews 2024, 124
(5)
, 2699-2804. https://doi.org/10.1021/acs.chemrev.3c00778
- Yang Yang, Xiaoyang Liu, Xiaofeng Wu, Gaolin Liang. Enzyme-Instructed CBT-Cys-like Click Cyclization Reactions for Bioimaging. Chemical & Biomedical Imaging 2024, 2
(2)
, 98-116. https://doi.org/10.1021/cbmi.3c00117
- Andrew M. Giltrap, Yizhi Yuan, Benjamin G. Davis. Late-Stage Functionalization of Living Organisms: Rethinking Selectivity in Biology. Chemical Reviews 2024, 124
(3)
, 889-928. https://doi.org/10.1021/acs.chemrev.3c00579
- Cinzia D’Avino, Sara Gutiérrez, Max J. Feldhaus, María Tomás-Gamasa, José Luis Mascareñas. Intracellular Synthesis of Indoles Enabled by Visible-Light Photocatalysis. Journal of the American Chemical Society 2024, 146
(5)
, 2895-2900. https://doi.org/10.1021/jacs.3c13647
- Amit Kumar, In Su Lee. Designer Nanoreactors for Bioorthogonal Catalysis. Accounts of Chemical Research 2024, 57
(3)
, 413-427. https://doi.org/10.1021/acs.accounts.3c00735
- Houqi Guo, Xin Ruan, Zekun Xu, Kai Wang, Xiang Li, Jun Jiang. Visible-Light-Mediated Dual Functionalization of Allenes: Regio- and Stereoselective Synthesis of Vinylsulfone Azides. The Journal of Organic Chemistry 2024, 89
(1)
, 665-675. https://doi.org/10.1021/acs.joc.3c02515
- Qingqiang Min, Xingyue Ji. Bioorthogonal Bond Cleavage Chemistry for On-demand Prodrug Activation: Opportunities and Challenges. Journal of Medicinal Chemistry 2023, 66
(24)
, 16546-16567. https://doi.org/10.1021/acs.jmedchem.3c01459
- Zhicheng Yan, Yiyao Pan, Guofeng Jiao, Mengyu Xu, Dongguang Fan, Ziwei Hu, Jiarui Wu, Tao Chen, Miao Liu, Xiaoguang Bao, Hengte Ke, Xingyue Ji. A Bioorthogonal Decaging Chemistry of N-Oxide and Silylborane for Prodrug Activation both In Vitro and In Vivo. Journal of the American Chemical Society 2023, 145
(45)
, 24698-24706. https://doi.org/10.1021/jacs.3c08012
- Giulia Mazzoccanti, Roberta Franzini, Alessia Ciogli, Marco Pierini, Fabio Buonsenso, Cristina Faggi, Antonio Francioso, Simone Manetto, Claudio Villani. Theoretically Predicted and Experimentally Detected Chirality of Dibenzocyclooctynes and Their Triazole Adducts with Azides. The Journal of Organic Chemistry 2023, 88
(21)
, 15097-15105. https://doi.org/10.1021/acs.joc.3c01557
- Chau Ngo, William Fried, Saba Aliyari, Joshua Feng, Chao Qin, Shilei Zhang, Hanjing Yang, Jean Shanaa, Pinghui Feng, Genhong Cheng, Xiaojiang S. Chen, Chao Zhang. Alkyne as a Latent Warhead to Covalently Target SARS-CoV-2 Main Protease. Journal of Medicinal Chemistry 2023, 66
(17)
, 12237-12248. https://doi.org/10.1021/acs.jmedchem.3c00810
- Athanasios Markos, Maurice Biedermann, Johannes Heimgärtner, Adeline Schmitt, Kathrin Lang, Helma Wennemers. Introducing Azomethine Imines to Chemical Biology: Bioorthogonal Reaction with Isonitriles. Journal of the American Chemical Society 2023, 145
(36)
, 19513-19517. https://doi.org/10.1021/jacs.3c07006
- Cunyuan Zhao, Yuankai Wang, Quan Pham, Changhang Dai, Abhishek Chatterjee, Masayuki Wasa. Chemical Tagging of Bioactive Amides by Cooperative Catalysis: Applications in the Syntheses of Drug Conjugates. Journal of the American Chemical Society 2023, 145
(26)
, 14233-14250. https://doi.org/10.1021/jacs.3c00169
- Yuanhang Xiang, Hui Zhang, Hao Lu, Binqi Wei, Cuiyun Su, Xiaojie Qin, Min Fang, Xinchun Li, Fan Yang. Bioorthogonal Microbubbles with Antifouling Nanofilm for Instant and Suspended Enrichment of Circulating Tumor Cells. ACS Nano 2023, 17
(10)
, 9633-9646. https://doi.org/10.1021/acsnano.3c03194
- Ming Fang, Gangam Srikanth Kumar, Stefano Racioppi, Heyang Zhang, Johnathan D. Rabb, Eva Zurek, Qing Lin. Hydrazonyl Sultones as Stable Tautomers of Highly Reactive Nitrile Imines for Fast Bioorthogonal Ligation Reaction. Journal of the American Chemical Society 2023, 145
(18)
, 9959-9964. https://doi.org/10.1021/jacs.2c12325
- Yuanyuan Li, Hongling Zhou, Huili Liu, Youfang Gan, Yaying Zheng, Jia Sheng, Rui Wang. Development of Nitroso-Based Probes for Labeling and Regulation of RAS Proteins in Cancer Cells via Sequential Ene-Ligation and Oxime Condensation. The Journal of Organic Chemistry 2023, 88
(3)
, 1762-1771. https://doi.org/10.1021/acs.joc.2c02922
- Kern Hast, M. Rhia L. Stone, Zhaojun Jia, Melih Baci, Tushar Aggarwal, Enver Cagri Izgu. Bioorthogonal Functionalization of Material Surfaces with Bioactive Molecules. ACS Applied Materials & Interfaces 2023, 15
(4)
, 4996-5009. https://doi.org/10.1021/acsami.2c20942
- Zhi-Chen Wu, Dale L. Boger. 1,2,3,5-Tetrazines: A General Synthesis, Cycloaddition Scope, and Fundamental Reactivity Patterns. The Journal of Organic Chemistry 2022, 87
(24)
, 16829-16846. https://doi.org/10.1021/acs.joc.2c02687
- Dongxia Chen, Yaying Lin, Ao Li, Xiangjie Luo, Chaoyong Yang, Jinhao Gao, Hongyu Lin. Bio-orthogonal Metabolic Fluorine Labeling Enables Deep-Tissue Visualization of Tumor Cells In Vivo by 19F Magnetic Resonance Imaging. Analytical Chemistry 2022, 94
(48)
, 16614-16621. https://doi.org/10.1021/acs.analchem.2c02443
- Natsumi Ueda, Shiho Sawada, Fumiya Yuasa, Karen Kato, Koji Nagahama. Covalent Stem Cell-Combining Injectable Materials with Enhanced Stemness and Controlled Differentiation In Vivo. ACS Applied Materials & Interfaces 2022, 14
(47)
, 52618-52633. https://doi.org/10.1021/acsami.2c12918
- Jianbing Wu, Yihui Hu, Hui Ye, Sheng Zhang, Jie Zhu, Duorui Ji, Yihua Zhang, Ya Ding, Zhangjian Huang. One Stone Two Birds: Redox-Sensitive Colocalized Delivery of Cisplatin and Nitric Oxide through Cascade Reactions. JACS Au 2022, 2
(10)
, 2339-2351. https://doi.org/10.1021/jacsau.2c00390
- Yayue Wang, Guohua Shen, Jie Li, Wuyu Mao, Hongbao Sun, Ping Feng, Haoxing Wu. Bioorthogonal Cleavage of Tetrazine-Caged Ethers and Esters Triggered by trans-Cyclooctene. Organic Letters 2022, 24
(29)
, 5293-5297. https://doi.org/10.1021/acs.orglett.2c01873
- Samira Sadeghi, Nihar D. Masurkar, Girish Vallerinteavide Mavelli, Siddharth Deshpande, Warren Kok Yong Tan, Sherman Yee, Shin-Ae Kang, Yoon-Pin Lim, Edward Kai-Hua Chow, Chester L. Drum. Bioorthogonal Catalysis for Treatment of Solid Tumors Using Thermostable, Self-Assembling, Single Enzyme Nanoparticles and Natural Product Conversion with Indole-3-acetic Acid. ACS Nano 2022, 16
(7)
, 10292-10301. https://doi.org/10.1021/acsnano.1c11560
- Rui Huang, Cristina-Maria Hirschbiegel, Xianzhi Zhang, Aarohi Gupta, Stefano Fedeli, Yisheng Xu, Vincent M. Rotello. Engineered Polymer-Supported Biorthogonal Nanocatalysts Using Flash Nanoprecipitation. ACS Applied Materials & Interfaces 2022, 14
(28)
, 31594-31600. https://doi.org/10.1021/acsami.2c04496
- Amit Dahiya, Franziska Schoenebeck. Orthogonal and Modular Arylation of Alkynylgermanes. ACS Catalysis 2022, 12
(13)
, 8048-8054. https://doi.org/10.1021/acscatal.2c02179
- Tyler K. Heiss, Robert S. Dorn, Andrew. J. Ferreira, Anna C. Love, Jennifer A. Prescher. Fluorogenic Cyclopropenones for Multicomponent, Real-Time Imaging. Journal of the American Chemical Society 2022, 144
(17)
, 7871-7880. https://doi.org/10.1021/jacs.2c02058
- Umberto M. Battisti, Rocío García-Vázquez, Dennis Svatunek, Barbara Herrmann, Andreas Löffler, Hannes Mikula, Matthias Manfred Herth. Synergistic Experimental and Computational Investigation of the Bioorthogonal Reactivity of Substituted Aryltetrazines. Bioconjugate Chemistry 2022, 33
(4)
, 608-624. https://doi.org/10.1021/acs.bioconjchem.2c00042
- Alessia Battigelli, Bethany Almeida, Anita Shukla. Recent Advances in Bioorthogonal Click Chemistry for Biomedical Applications. Bioconjugate Chemistry 2022, 33
(2)
, 263-271. https://doi.org/10.1021/acs.bioconjchem.1c00564
- Guopan Liu, Ming Ho Choi, Haiying Ma, Xuejiao Guo, Pui-Chi Lo, Jinyong Kim, Liang Zhang. Bioorthogonal Conjugation-Assisted Purification Method for Profiling Cell Surface Proteome. Analytical Chemistry 2022, 94
(3)
, 1901-1909. https://doi.org/10.1021/acs.analchem.1c05187
- Gangam Srikanth Kumar, Stefano Racioppi, Eva Zurek, Qing Lin. Superfast Tetrazole–BCN Cycloaddition Reaction for Bioorthogonal Protein Labeling on Live Cells. Journal of the American Chemical Society 2022, 144
(1)
, 57-62. https://doi.org/10.1021/jacs.1c10354
- Angus S. Mackay, Richard J. Payne, Lara R. Malins. Electrochemistry for the Chemoselective Modification of Peptides and Proteins. Journal of the American Chemical Society 2022, 144
(1)
, 23-41. https://doi.org/10.1021/jacs.1c11185
- Shahar Tevet, Shreyas S. Wagle, Gadi Slor, Roey J. Amir. Tuning the Reactivity of Micellar Nanoreactors by Precise Adjustments of the Amphiphile and Substrate Hydrophobicity. Macromolecules 2021, 54
(24)
, 11419-11426. https://doi.org/10.1021/acs.macromol.1c01755
- Robert E. Bird, Steven A. Lemmel, Xiang Yu, Qiongqiong Angela Zhou. Bioorthogonal Chemistry and Its Applications. Bioconjugate Chemistry 2021, 32
(12)
, 2457-2479. https://doi.org/10.1021/acs.bioconjchem.1c00461
- Fa-Guang Zhang, Zhen Chen, Xiaodong Tang, Jun-An Ma. Triazines: Syntheses and Inverse Electron-demand Diels–Alder Reactions. Chemical Reviews 2021, 121
(23)
, 14555-14593. https://doi.org/10.1021/acs.chemrev.1c00611
- Anna Kataki-Anastasakou, Selena Hernandez, Ellen M. Sletten. Cell-surface Labeling via Bioorthogonal Host–Guest Chemistry. ACS Chemical Biology 2021, 16
(11)
, 2124-2129. https://doi.org/10.1021/acschembio.1c00494
- Umberto M. Battisti, Klas Bratteby, Jesper T. Jørgensen, Lars Hvass, Vladimir Shalgunov, Hannes Mikula, Andreas Kjær, Matthias Manfred Herth. Development of the First Aliphatic 18F-Labeled Tetrazine Suitable for Pretargeted PET Imaging—Expanding the Bioorthogonal Tool Box. Journal of Medicinal Chemistry 2021, 64
(20)
, 15297-15312. https://doi.org/10.1021/acs.jmedchem.1c01326
- Carolina Carrillo-Carrión, Raquel Martínez, Ester Polo, María Tomás-Gamasa, Paolo Destito, Manuel Ceballos, Beatriz Pelaz, Fernando López, José L. Mascareñas, Pablo del Pino. Plasmonic-Assisted Thermocyclizations in Living Cells Using Metal–Organic Framework Based Nanoreactors. ACS Nano 2021, 15
(10)
, 16924-16933. https://doi.org/10.1021/acsnano.1c07983
- Suhail A. Rather, Mohammad Yaqoob Bhat, Feroze Hussain, Qazi Naveed Ahmed. Sulfonyl-Promoted Michaelis–Arbuzov-Type Reaction: An Approach to S/Se–P Bonds. The Journal of Organic Chemistry 2021, 86
(19)
, 13644-13663. https://doi.org/10.1021/acs.joc.1c01681
- Christopher J. Addonizio, Brant D. Gates, Matthew J. Webber. Supramolecular “Click Chemistry” for Targeting in the Body. Bioconjugate Chemistry 2021, 32
(9)
, 1935-1946. https://doi.org/10.1021/acs.bioconjchem.1c00326
- Aaditya Govindarajan, Vasuki Gnanasambandam. Toward Intracellular Bioconjugation Using Transition-Metal-Free Techniques. Bioconjugate Chemistry 2021, 32
(8)
, 1431-1454. https://doi.org/10.1021/acs.bioconjchem.1c00173
- Trang Huyen Le-Kim, Bon Il Koo, Sung Duk Jo, Nai-Wen Liang, Moon Young Yang, In Cho, Jae Byum Chang, Tzu-Wei Wang, Yoon Sung Nam. Artificial Taste Buds: Bioorthogonally Ligated Gustatory–Neuronal Multicellular Hybrids Enabling Intercellular Taste Signal Transmission. ACS Biomaterials Science & Engineering 2021, 7
(8)
, 3783-3792. https://doi.org/10.1021/acsbiomaterials.1c00247
- Chuanqi Wang, He Zhang, Tao Zhang, Xiaoyu Zou, Hui Wang, Julia E. Rosenberger, Raghu Vannam, William S. Trout, Jonathan B. Grimm, Luke D. Lavis, Colin Thorpe, Xinqiao Jia, Zibo Li, Joseph M. Fox. Enabling In Vivo Photocatalytic Activation of Rapid Bioorthogonal Chemistry by Repurposing Silicon-Rhodamine Fluorophores as Cytocompatible Far-Red Photocatalysts. Journal of the American Chemical Society 2021, 143
(28)
, 10793-10803. https://doi.org/10.1021/jacs.1c05547
- Mohammad Shafee Alkanawati, Marina Machtakova, Katharina Landfester, Héloïse Thérien-Aubin. Bio-Orthogonal Nanogels for Multiresponsive Release. Biomacromolecules 2021, 22
(7)
, 2976-2984. https://doi.org/10.1021/acs.biomac.1c00378
- Didier A. Bilodeau, Kaitlyn D. Margison, Mariam Serhan, John Paul Pezacki. Bioorthogonal Reactions Utilizing Nitrones as Versatile Dipoles in Cycloaddition Reactions. Chemical Reviews 2021, 121
(12)
, 6699-6717. https://doi.org/10.1021/acs.chemrev.0c00832
- Titas Deb, Julian Tu, Raphael M. Franzini. Mechanisms and Substituent Effects of Metal-Free Bioorthogonal Reactions. Chemical Reviews 2021, 121
(12)
, 6850-6914. https://doi.org/10.1021/acs.chemrev.0c01013
- Brian J. Levandowski, Ronald T. Raines. Click Chemistry with Cyclopentadiene. Chemical Reviews 2021, 121
(12)
, 6777-6801. https://doi.org/10.1021/acs.chemrev.0c01055
- Tyler K. Heiss, Robert S. Dorn, Jennifer A. Prescher. Bioorthogonal Reactions of Triarylphosphines and Related Analogues. Chemical Reviews 2021, 121
(12)
, 6802-6849. https://doi.org/10.1021/acs.chemrev.1c00014
- Jie Wang, Xin Wang, Xinyuan Fan, Peng R. Chen. Unleashing the Power of Bond Cleavage Chemistry in Living Systems. ACS Central Science 2021, 7
(6)
, 929-943. https://doi.org/10.1021/acscentsci.1c00124
- Amol D. Pagar, Mahesh D. Patil, Dillon T. Flood, Tae Hyeon Yoo, Philip E. Dawson, Hyungdon Yun. Recent Advances in Biocatalysis with Chemical Modification and Expanded Amino Acid Alphabet. Chemical Reviews 2021, 121
(10)
, 6173-6245. https://doi.org/10.1021/acs.chemrev.0c01201
- Zhiguo Gao, Yaojia Li, Zhikun Liu, Yu Zhang, Fanghui Chen, Peijing An, Wenjun Lu, Jinzhong Hu, Chaoqun You, Jun Xu, Xiangyang Zhang, Baiwang Sun. Small-Molecule-Selective Organosilica Nanoreactors for Copper-Catalyzed Azide–Alkyne Cycloaddition Reactions in Cellular and Living Systems. Nano Letters 2021, 21
(8)
, 3401-3409. https://doi.org/10.1021/acs.nanolett.0c04930
- Al-Mahmnur Alam, Young-Seok Shon. Water-Soluble Noble Metal Nanoparticle Catalysts Capped with Small Organic Molecules for Organic Transformations in Water. ACS Applied Nano Materials 2021, 4
(4)
, 3294-3318. https://doi.org/10.1021/acsanm.1c00335
- Dahye Kang, Justin Kim. Bioorthogonal Retro-Cope Elimination Reaction of N,N-Dialkylhydroxylamines and Strained Alkynes. Journal of the American Chemical Society 2021, 143
(15)
, 5616-5621. https://doi.org/10.1021/jacs.1c00885
- Chandra Kanth Bandi, Kyle S. Skalenko, Ayushi Agrawal, Neelan Sivaneri, Margaux Thiry, Shishir P. S. Chundawat. Engineered Regulon to Enable Autonomous Azide Ion Biosensing, Recombinant Protein Production, and in Vivo Glycoengineering. ACS Synthetic Biology 2021, 10
(4)
, 682-689. https://doi.org/10.1021/acssynbio.0c00449
- Xiangzhao Ai, Shuyan Wang, Yaou Duan, Qiangzhe Zhang, Maggie S. Chen, Weiwei Gao, Liangfang Zhang. Emerging Approaches to Functionalizing Cell Membrane-Coated Nanoparticles. Biochemistry 2021, 60
(13)
, 941-955. https://doi.org/10.1021/acs.biochem.0c00343
- Andrea Peier, Lan Ge, Nicolas Boyer, John Frost, Ruchia Duggal, Kaustav Biswas, Scott Edmondson, Jeffrey D. Hermes, Lin Yan, Chad Zimprich, Ahmad Sadruddin, Hung Yi Kristal Kaan, Arun Chandramohan, Christopher J. Brown, Dawn Thean, Xue Er Lee, Tsz Ying Yuen, Fernando J. Ferrer-Gago, Charles W. Johannes, David P. Lane, Brad Sherborne, Cesear Corona, Matthew B. Robers, Tomi K. Sawyer, Anthony W. Partridge. NanoClick: A High Throughput, Target-Agnostic Peptide Cell Permeability Assay. ACS Chemical Biology 2021, 16
(2)
, 293-309. https://doi.org/10.1021/acschembio.0c00804
- Zhigang Lyu, Yue Zhao, Zakey Yusuf Buuh, Nicole Gorman, Aaron R. Goldman, Md Shafiqul Islam, Hsin-Yao Tang, Rongsheng E. Wang. Steric-Free Bioorthogonal Labeling of Acetylation Substrates Based on a Fluorine–Thiol Displacement Reaction. Journal of the American Chemical Society 2021, 143
(3)
, 1341-1347. https://doi.org/10.1021/jacs.0c05605
- Vera F. C. Ferreira, Bruno L. Oliveira, Alice D’Onofrio, Carlos M. Farinha, Lurdes Gano, António Paulo, Gonçalo J. L. Bernardes, Filipa Mendes. In Vivo Pretargeting Based on Cysteine-Selective Antibody Modification with IEDDA Bioorthogonal Handles for Click Chemistry. Bioconjugate Chemistry 2021, 32
(1)
, 121-132. https://doi.org/10.1021/acs.bioconjchem.0c00551
- Jessica Hersh, David Broyles, José Manuel Condor Capcha, Emre Dikici, Lina A. Shehadeh, Sylvia Daunert, Sapna Deo. Peptide-Modified Biopolymers for Biomedical Applications. ACS Applied Bio Materials 2021, 4
(1)
, 229-251. https://doi.org/10.1021/acsabm.0c01145
- Yichuan Zhang, Muhammed Üçüncü, Alessia Gambardella, Assel Baibek, Jin Geng, Shuo Zhang, Jessica Clavadetscher, Inga Litzen, Mark Bradley, Annamaria Lilienkampf. Bioorthogonal Swarming: In Situ Generation of Dendrimers. Journal of the American Chemical Society 2020, 142
(52)
, 21615-21621. https://doi.org/10.1021/jacs.0c07869
- Rajdip Chowdhury, Zhunzhun Yu, My Linh Tong, Stefanie V. Kohlhepp, Xiang Yin, Abraham Mendoza. Decarboxylative Alkyl Coupling Promoted by NADH and Blue Light. Journal of the American Chemical Society 2020, 142
(47)
, 20143-20151. https://doi.org/10.1021/jacs.0c09678
- Tao Zhang, Jianhua Cai, Hui Wang, Mengzhe Wang, Hong Yuan, Zhanhong Wu, Xiaofen Ma, Zibo Li. RXH-Reactive 18F-Vinyl Sulfones as Versatile Agents for PET Probe Construction. Bioconjugate Chemistry 2020, 31
(11)
, 2482-2487. https://doi.org/10.1021/acs.bioconjchem.0c00487
- Martin Wilkovitsch, Maximilian Haider, Barbara Sohr, Barbara Herrmann, Jenna Klubnick, Ralph Weissleder, Jonathan C. T. Carlson, Hannes Mikula. A Cleavable C2-Symmetric trans-Cyclooctene Enables Fast and Complete Bioorthogonal Disassembly of Molecular Probes. Journal of the American Chemical Society 2020, 142
(45)
, 19132-19141. https://doi.org/10.1021/jacs.0c07922
- Yun Hu, Jessica M. Roberts, Henry R. Kilgore, Amirah S. Mat Lani, Ronald T. Raines, Jennifer M. Schomaker. Triple, Mutually Orthogonal Bioorthogonal Pairs through the Design of Electronically Activated Sulfamate-Containing Cycloalkynes. Journal of the American Chemical Society 2020, 142
(44)
, 18826-18835. https://doi.org/10.1021/jacs.0c06725
- Dillon T. Flood, Kyle W. Knouse, Julien C. Vantourout, Seiya Kitamura, Brittany B. Sanchez, Emily J. Sturgell, Jason S. Chen, Dennis W. Wolan, Phil S. Baran, Philip E. Dawson. Synthetic Elaboration of Native DNA by RASS (SENDR). ACS Central Science 2020, 6
(10)
, 1789-1799. https://doi.org/10.1021/acscentsci.0c00680
- Sandeep Palvai, Jasmine Bhangu, Burcin Akgun, Christopher T. Moody, Dennis G. Hall, Yevgeny Brudno. In Vivo Targeting Using Arylboronate/Nopoldiol Click Conjugation. Bioconjugate Chemistry 2020, 31
(10)
, 2288-2292. https://doi.org/10.1021/acs.bioconjchem.0c00453
- Yanira Méndez, Aldrin V. Vasco, Ana R. Humpierre, Bernhard Westermann. Isonitriles: Versatile Handles for the Bioorthogonal Functionalization of Proteins. ACS Omega 2020, 5
(40)
, 25505-25510. https://doi.org/10.1021/acsomega.0c03728
- Jerrin Thomas George, Mohd. Azhar, Meghali Aich, Dipanjali Sinha, Uddhav B. Ambi, Souvik Maiti, Debojyoti Chakraborty, Seergazhi G. Srivatsan. Terminal Uridylyl Transferase Mediated Site-Directed Access to Clickable Chromatin Employing CRISPR-dCas9. Journal of the American Chemical Society 2020, 142
(32)
, 13954-13965. https://doi.org/10.1021/jacs.0c06541
- Julia Moschny, Wolfram Lorenzen, Alexandra Hilfer, Robert Eckenstaler, Stefan Jahns, Heike Enke, Dan Enke, Philipp Schneider, Ralf A. Benndorf, Timo H. J. Niedermeyer. Precursor-Directed Biosynthesis and Fluorescence Labeling of Clickable Microcystins. Journal of Natural Products 2020, 83
(6)
, 1960-1970. https://doi.org/10.1021/acs.jnatprod.0c00251
- Miguel Macias-Contreras, Huan He, Kevin N. Little, Justin P. Lee, Ryan P. Campbell, Maksim Royzen, Lei Zhu. SNAP/CLIP-Tags and Strain-Promoted Azide–Alkyne Cycloaddition (SPAAC)/Inverse Electron Demand Diels–Alder (IEDDA) for Intracellular Orthogonal/Bioorthogonal Labeling. Bioconjugate Chemistry 2020, 31
(5)
, 1370-1381. https://doi.org/10.1021/acs.bioconjchem.0c00107
- Jordi Agramunt, Rebecca Ginesi, Enrique Pedroso, Anna Grandas. Inverse Electron-Demand Diels–Alder Bioconjugation Reactions Using 7-Oxanorbornenes as Dienophiles. The Journal of Organic Chemistry 2020, 85
(10)
, 6593-6604. https://doi.org/10.1021/acs.joc.0c00583
- Eduardo Ruivo, Filipe Elvas, Karuna Adhikari, Christel Vangestel, Glenn Van Haesendonck, Filip Lemière, Steven Staelens, Sigrid Stroobants, Pieter Van der Veken, Leonie wyffels, Koen Augustyns. Preclinical Evaluation of a Novel 18F-Labeled dTCO-Amide Derivative for Bioorthogonal Pretargeted Positron Emission Tomography Imaging. ACS Omega 2020, 5
(9)
, 4449-4456. https://doi.org/10.1021/acsomega.9b03584
- Wenqi Yu, Rui Liu, Yang Zhou, Huile Gao. Size-Tunable Strategies for a Tumor Targeted Drug Delivery System. ACS Central Science 2020, 6
(2)
, 100-116. https://doi.org/10.1021/acscentsci.9b01139
- Mary R. Adams, Christopher T. Moody, Jennifer L. Sollinger, Yevgeny Brudno. Extracellular-Matrix-Anchored Click Motifs for Specific Tissue Targeting. Molecular Pharmaceutics 2020, 17
(2)
, 392-403. https://doi.org/10.1021/acs.molpharmaceut.9b00589
- M. Soledad Garre, Raúl Losantos, Sara Gutiérrez, David Sucunza, Patricia García-García, Diego Sampedro, Juan J. Vaquero. Synthesis and Photophysical Behavior of a Highly Fluorescent Family of Unsymmetrical Organoboron Complexes Containing 5-(Pyridin-2-ylmethylene)imidazolidine-2,4-dione Moieties. The Journal of Organic Chemistry 2020, 85
(2)
, 441-448. https://doi.org/10.1021/acs.joc.9b02451
- Suttipong Suttapitugsakul, Fangxu Sun, Ronghu Wu. Recent Advances in Glycoproteomic Analysis by Mass Spectrometry. Analytical Chemistry 2020, 92
(1)
, 267-291. https://doi.org/10.1021/acs.analchem.9b04651
- Shuang Song Kaitlyn Ngo Kathryn Uhrich . Green Chemistry Principles In Advancing Hierarchical Functionalization of Polymer-Based Nanomedicines. 2020, 135-157. https://doi.org/10.1021/bk-2020-1372.ch008
- Yulin Tian, Qing Lin. Fitness Factors for Bioorthogonal Chemical Probes. ACS Chemical Biology 2019, 14
(12)
, 2489-2496. https://doi.org/10.1021/acschembio.9b00755
- Rebecca J. B. Schäfer, Mattia R. Monaco, Mao Li, Alina Tirla, Pablo Rivera-Fuentes, Helma Wennemers. The Bioorthogonal Isonitrile–Chlorooxime Ligation. Journal of the American Chemical Society 2019, 141
(47)
, 18644-18648. https://doi.org/10.1021/jacs.9b07632
- Gaochan Wu, Tong Zhao, Dongwei Kang, Jian Zhang, Yuning Song, Vigneshwaran Namasivayam, Jacob Kongsted, Christophe Pannecouque, Erik De Clercq, Vasanthanathan Poongavanam, Xinyong Liu, Peng Zhan. Overview of Recent Strategic Advances in Medicinal Chemistry. Journal of Medicinal Chemistry 2019, 62
(21)
, 9375-9414. https://doi.org/10.1021/acs.jmedchem.9b00359
- Valerio Sabatino, Johannes G. Rebelein, Thomas R. Ward. “Close-to-Release”: Spontaneous Bioorthogonal Uncaging Resulting from Ring-Closing Metathesis. Journal of the American Chemical Society 2019, 141
(43)
, 17048-17052. https://doi.org/10.1021/jacs.9b07193
- Sen Yang, Shuangyu Zhu, Dengfu Lu, Yuefa Gong. Polarity-Reversed Addition of Enol Ethers to Imines under Visible Light: Redox-Neutral Access to Azide-Containing Amino Acids. Organic Letters 2019, 21
(20)
, 8464-8468. https://doi.org/10.1021/acs.orglett.9b03238
- Han Gu, Saptarshi Ghosh, Richard J. Staples, Susan L. Bane. β-Hydroxy-Stabilized Boron–Nitrogen Heterocycles Enable Rapid and Efficient C-Terminal Protein Modification. Bioconjugate Chemistry 2019, 30
(10)
, 2604-2613. https://doi.org/10.1021/acs.bioconjchem.9b00534
- Evan B. Glass, Shirin Masjedi, Stephanie O. Dudzinski, Andrew J. Wilson, Craig L. Duvall, Fiona E. Yull, Todd D. Giorgio. Optimizing Mannose “Click” Conjugation to Polymeric Nanoparticles for Targeted siRNA Delivery to Human and Murine Macrophages. ACS Omega 2019, 4
(16)
, 16756-16767. https://doi.org/10.1021/acsomega.9b01465
- Elena Petit, Lluís Bosch, Anna M. Costa, Jaume Vilarrasa. (Z)-Oxopropene-1,3-diyl, a Linker for the Conjugation of the Thiol Group of Cysteine with Amino-Derivatized Drugs. The Journal of Organic Chemistry 2019, 84
(17)
, 11170-11176. https://doi.org/10.1021/acs.joc.8b02686
- Ulrike Reisacher, Bastian Groitl, Ralf Strasser, Gergely B. Cserép, Péter Kele, Hans-Achim Wagenknecht. Triazine-Modified 7-Deaza-2′-deoxyadenosines: Better Suited for Bioorthogonal Labeling of DNA by PCR than 2′-Deoxyuridines. Bioconjugate Chemistry 2019, 30
(6)
, 1773-1780. https://doi.org/10.1021/acs.bioconjchem.9b00295
- Tyler
K. Heiss, Jennifer A. Prescher. Cyclopropeniminium Ions Exhibit Unique Reactivity Profiles with Bioorthogonal Phosphines. The Journal of Organic Chemistry 2019, 84
(11)
, 7443-7448. https://doi.org/10.1021/acs.joc.9b00518
- Yuanwei Dai, Jiaping Weng, Justin George, Huan Chen, Qishan Lin, Jun Wang, Maksim Royzen, Qiang Zhang. Three-Component Protein Modification Using Mercaptobenzaldehyde Derivatives. Organic Letters 2019, 21
(10)
, 3828-3833. https://doi.org/10.1021/acs.orglett.9b01294
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 70 other publications.
- 1Hang, H. C.; Yu, C.; Kato, D. L.; Bertozzi, C. R. A metabolic labeling approach toward proteomic analysis of mucin-type O-linked glycosylation. Proc. Natl. Acad. Sci. U. S. A. 2003, 100, 14846– 14851, DOI: 10.1073/pnas.23352011001https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXpvFaqsbk%253D&md5=225b7d0f94f9684e4595c68b725d3425A metabolic labeling approach toward proteomic analysis of mucin-type O-linked glycosylationHang, Howard C.; Yu, Chong; Kato, Darryl L.; Bertozzi, Carolyn R.Proceedings of the National Academy of Sciences of the United States of America (2003), 100 (25), 14846-14851CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Mucin-type O-linked glycoproteins are involved in a variety of biol. interactions in higher eukaryotes. The biosynthesis of these glycoproteins is initiated by a family of polypeptide N-acetyl-α-galactosaminyltransferases (ppGalNAcTs) that modify proteins in the secretory pathway. The lack of a defined consensus sequence for the ppGalNAcTs makes the prediction of mucin-type O-linked glycosylation difficult based on primary sequence alone. Herein we present a method for labeling mucin-type O-linked glycoproteins with a unique chem. tag, the azide, which permits their selective covalent modification from complex cell lysates. From a panel of synthetic derivs., we identified an azido GalNAc analog (N-azidoacetylgalactosamine, GalNAz) that is metabolized by numerous cell types and installed on mucin-type O-linked glycoproteins by the ppGalNAcTs. The azide serves as a bioorthogonal chem. handle for selective modification with biochem. or biophys. probes using the Staudinger ligation. The approach was validated by labeling a recombinant glycoprotein that is known to possess O-linked glycans with GalNAz. In addn., GalNAz efficiently labeled mucin-type O-linked glycoproteins expressed at endogenous levels. The ability to label mucin-type O-linked glycoproteins with chem. tags should facilitate their identification by proteomic strategies.
- 2Bertozzi, C. R. A decade of bioorthogonal chemistry. Acc. Chem. Res. 2011, 44, 651– 653, DOI: 10.1021/ar200193f2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtFOitrvN&md5=d864c8ac9684dfd32f6c352669d49fdbA Decade of Bioorthogonal ChemistryBertozzi, Carolyn R.Accounts of Chemical Research (2011), 44 (9), 651-653CODEN: ACHRE4; ISSN:0001-4842. (American Chemical Society)There is no expanded citation for this reference.
- 3Sletten, E. M.; Bertozzi, C. R. Bioorthogonal chemistry: fishing for selectivity in a sea of functionality. Angew. Chem., Int. Ed. 2009, 48, 6974– 6998, DOI: 10.1002/anie.2009009423https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtV2ltrvK&md5=993199cb4deabc186848bae6fe81f605Bioorthogonal Chemistry: Fishing for Selectivity in a Sea of FunctionalitySletten, Ellen M.; Bertozzi, Carolyn R.Angewandte Chemie, International Edition (2009), 48 (38), 6974-6998CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. The study of biomols. in their native environments is a challenging task because of the vast complexity of cellular systems. Technologies developed in the last few years for the selective modification of biol. species in living systems have yielded new insights into cellular processes. Key to these new techniques are bioorthogonal chem. reactions, whose components must react rapidly and selectively with each other under physiol. conditions in the presence of the plethora of functionality necessary to sustain life. Herein the authors describe the bioorthogonal chem. reactions developed to date and how they can be used to study biomols.
- 4Baskin, J. M.; Prescher, J. A.; Laughlin, S. T.; Agard, N. J.; Chang, P. V.; Miller, I. A.; Lo, A.; Codelli, J. A.; Bertozzi, C. R. Copper-free click chemistry for dynamic in vivo imaging. Proc. Natl. Acad. Sci. U. S. A. 2007, 104, 16793– 16797, DOI: 10.1073/pnas.07070901044https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXht1KgtLvN&md5=7ea429c09b86509ff7f3960a95f11276Copper-free click chemistry for dynamic in vivo imagingBaskin, Jeremy M.; Prescher, Jennifer A.; Laughlin, Scott T.; Agard, Nicholas J.; Chang, Pamela V.; Miller, Isaac A.; Lo, Anderson; Codelli, Julian A.; Bertozzi, Carolyn R.Proceedings of the National Academy of Sciences of the United States of America (2007), 104 (43), 16793-16797CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Dynamic imaging of proteins in live cells is routinely performed by using genetically encoded reporters, an approach that cannot be extended to other classes of biomols. such as glycans and lipids. Here, the authors report a Cu-free variant of click chem. that can label these biomols. rapidly and selectively in living systems, overcoming the intrinsic toxicity of the canonical Cu-catalyzed reaction. The crit. reagent, a substituted cyclooctyne, possesses ring strain and electron-withdrawing fluorine substituents that together promote the [3+2] dipolar cycloaddn. with azides installed metabolically into biomols. This Cu-free click reaction possesses comparable kinetics to the Cu-catalyzed reaction and proceeds within minutes on live cells with no apparent toxicity. With this technique, the authors studied the dynamics of glycan trafficking and identified a population of sialoglycoconjugates with unexpectedly rapid internalization kinetics.
- 5Laughlin, S. T.; Baskin, J. M.; Amacher, S. L.; Bertozzi, C. R. In vivo imaging of membrane-associated glycans in developing zebrafish. Science 2008, 320, 664– 667, DOI: 10.1126/science.11551065https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXltFyis78%253D&md5=2f0a4dbef1ef18bba45013e3847387baIn Vivo Imaging of Membrane-Associated Glycans in Developing ZebrafishLaughlin, Scott T.; Baskin, Jeremy M.; Amacher, Sharon L.; Bertozzi, Carolyn R.Science (Washington, DC, United States) (2008), 320 (5876), 664-667CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Glycans are attractive targets for mol. imaging but have been inaccessible because of their incompatibility with genetically encoded reporters. We demonstrated the noninvasive imaging of glycans in live developing zebrafish, using a chem. reporter strategy. Zebrafish embryos were treated with an unnatural sugar to metabolically label their cell-surface glycans with azides. Subsequently, the embryos were reacted with fluorophore conjugates by means of copper-free click chem., enabling the visualization of glycans in vivo at subcellular resoln. during development. At 60 h after fertilization, we obsd. an increase in de novo glycan biosynthesis in the jaw region, pectoral fins, and olfactory organs. Using a multicolor detection strategy, we performed a spatiotemporal anal. of glycan expression and trafficking and identified patterns that would be undetectable with conventional mol. imaging approaches.
- 6Agarwal, P.; Beahm, B. J.; Shieh, P.; Bertozzi, C. R. Systemic Fluorescence Imaging of Zebrafish Glycans with Bioorthogonal Chemistry. Angew. Chem., Int. Ed. 2015, 54, 11504– 11510, DOI: 10.1002/anie.2015042496https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht1Kiu7vL&md5=454dc0471cc02051439df43c3fe4d55cSystemic Fluorescence Imaging of Zebrafish Glycans with Bioorthogonal ChemistryAgarwal, Paresh; Beahm, Brendan J.; Shieh, Peyton; Bertozzi, Carolyn R.Angewandte Chemie, International Edition (2015), 54 (39), 11504-11510CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Vertebrate glycans constitute a large, important, and dynamic set of post-translational modifications that are notoriously difficult to manipulate and image. Although the chem. reporter strategy has been used in conjunction with bioorthogonal chem. to image the external glycosylation state of live zebrafish and detect tumor-assocd. glycans in mice, the ability to image glycans systemically within a live organism has remained elusive. Here, the authors report a method that combines the metabolic incorporation of a cyclooctyne-functionalized sialic acid deriv. with a ligation reaction of a fluorogenic tetrazine, allowing for the imaging of sialylated glycoconjugates within live zebrafish embryos.
- 7Oller-Salvia, B.; Kym, G.; Chin, J. W. Rapid and Efficient Generation of Stable Antibody-Drug Conjugates via an Encoded Cyclopropene and an Inverse-Electron-Demand Diels-Alder Reaction. Angew. Chem., Int. Ed. 2018, 57, 2831– 2834, DOI: 10.1002/anie.2017123707https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXis1WjtLg%253D&md5=f2b16b8bdc875d4d0de3b32e649c2e67Rapid and Efficient Generation of Stable Antibody-Drug Conjugates via an Encoded Cyclopropene and an Inverse-Electron-Demand Diels-Alder ReactionOller-Salvia, Benjami; Kym, Gene; Chin, Jason W.Angewandte Chemie, International Edition (2018), 57 (11), 2831-2834CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Homogeneous antibody-drug conjugates (ADCs), generated by site-specific toxin linkage, show improved therapeutic indexes with respect to traditional ADCs. However, current methods to produce site-specific conjugates suffer from low protein expression, slow reaction kinetics, and low yields, or are limited to particular conjugation sites. Here we describe high yielding expression systems that efficiently incorporate a cyclopropene deriv. of lysine (CypK) into antibodies through genetic-code expansion. We express trastuzumab bearing CypK and conjugate tetrazine derivs. to the antibody. We show that the dihydropyridazine linkage resulting from the conjugation reaction is stable in serum, and generate an ADC bearing monomethyl auristatin E that selectively kills cells expressing a high level of HER2. Our results demonstrate that CypK is a minimal bioorthogonal handle for the rapid prodn. of stable therapeutic protein conjugates.
- 8Devaraj, N. K.; Thurber, G. M.; Keliher, E. J.; Marinelli, B.; Weissleder, R. Reactive polymer enables efficient in vivo bioorthogonal chemistry. Proc. Natl. Acad. Sci. U. S. A. 2012, 109, 4762– 4767, DOI: 10.1073/pnas.11134661098https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XltF2msbs%253D&md5=1e747e0637e4f98c48d2204321b8c4efReactive polymer enables efficient in vivo bioorthogonal chemistryDevaraj, Neal K.; Thurber, Greg M.; Keliher, Edmund J.; Marinelli, Brett; Weissleder, RalphProceedings of the National Academy of Sciences of the United States of America (2012), 109 (13), 4762-4767, S4762/1-S4762/2CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)There has been intense interest in the development of selective bioorthogonal reactions or "click" chem. that can proceed in live animals. Until now however, most reactions still require vast surpluses of reactants because of steep temporal and spatial concn. gradients. Using computational modeling and design of pharmacokinetically optimized reactants, we have developed a predictable method for efficient in vivo click reactions. Specifically, we show that polymer modified tetrazines (PMT) are a key enabler for in vivo bioorthogonal chem. based on the very fast and catalyst-free [4 + 2] tetrazine/trans-cyclooctene cycloaddn. Using fluorescent PMT for cellular resoln. and 18F labeled PMT for whole animal imaging, we show that cancer cell epitopes can be easily reacted in vivo. This generic strategy should help guide the design of future chemistries and find widespread use for different in vivo bioorthogonal applications, particularly in the biomedical sciences.
- 9Antonow, D. Fragment-based approaches and the prospect of fragmented prodrugs. Drug Discovery Today 2010, 15, 801– 803, DOI: 10.1016/j.drudis.2010.08.0159https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3cfot1GhtA%253D%253D&md5=af44714f2d9ed7711cede8a917f2d071Fragment-based approaches and the prospect of fragmented prodrugsAntonow DyeisonDrug discovery today (2010), 15 (19-20), 801-3 ISSN:.There is no expanded citation for this reference.
- 10Zengeya, T. T.; Garlick, J. M.; Kulkarni, R. A.; Miley, M.; Roberts, A. M.; Yang, Y.; Crooks, D. R.; Sourbier, C.; Linehan, W. M.; Meier, J. L. Co-opting a Bioorthogonal Reaction for Oncometabolite Detection. J. Am. Chem. Soc. 2016, 138, 15813– 15816, DOI: 10.1021/jacs.6b0970610https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvVygsr7N&md5=1476ba65466917dffe27c6e137f62c1cCo-opting a Bioorthogonal Reaction for Oncometabolite DetectionZengeya, Thomas T.; Garlick, Julie M.; Kulkarni, Rhushikesh A.; Miley, Mikayla; Roberts, Allison M.; Yang, Youfeng; Crooks, Daniel R.; Sourbier, Carole; Linehan, W. Marston; Meier, Jordan L.Journal of the American Chemical Society (2016), 138 (49), 15813-15816CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Dysregulated metab. is a hallmark of many diseases, including cancer. Methods to fluorescently detect metabolites have the potential to enable new approaches to cancer detection and imaging. However, fluorescent sensing methods for naturally occurring cellular metabolites are relatively unexplored. Here the authors report the development of a chem. approach to detect the oncometabolite fumarate. The authors' strategy exploits a known bioorthogonal reaction, the 1,3-dipolar cycloaddn. of nitrileimines and electron-poor olefins, to detect fumarate via fluorescent pyrazoline cycloadduct formation. The authors demonstrate that hydrazonyl chlorides serve as readily accessible nitrileimine precursors, whose reactivity and spectral properties can be tuned to enable detection of fumarate and other dipolarophile metabolites. Finally, the authors show this reaction can be used to detect enzyme activity changes caused by mutations in fumarate hydratase, which underlie the familial cancer predisposition syndrome hereditary leiomyomatosis and renal cell cancer. The authors' studies define a novel intersection of bioorthogonal chem. and metabolite reactivity that may be harnessed to enable biol. profiling, imaging, and diagnostic applications.
- 11Patterson, D. M.; Prescher, J. A. Orthogonal bioorthogonal chemistries. Curr. Opin. Chem. Biol. 2015, 28, 141– 149, DOI: 10.1016/j.cbpa.2015.07.00611https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtlWlsb7J&md5=80c1ef9d29769961cf4f17fbf21d9aa4Orthogonal bioorthogonal chemistriesPatterson, David M.; Prescher, Jennifer A.Current Opinion in Chemical Biology (2015), 28 (), 141-149CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)A review. Bioorthogonal reactions have long been used to examine individual biomols. in living systems. Studies of multi-component networks demand not only reliable bioorthogonal chemistries, but also combinations of reactions that can be used in tandem. Such orthogonal bioorthogonal transformations have been reported in recent years, and these chemistries are enabling new explorations into biol. This article highlights the development of orthogonal bioorthogonal reactions and their application in multi-target imaging and macromol. assembly. Methods to tune and control orthogonal reactivity are also discussed, along with prospects for identifying new classes of compatible reactions.
- 12Yu, J.; Shen, D.; Zhang, H.; Yin, Z. Rapid, Stoichiometric, Site-Specific Modification of Aldehyde-Containing Proteins Using a Tandem Knoevenagel-Intra Michael Addition Reaction. Bioconjugate Chem. 2018, 29, 1016– 1020, DOI: 10.1021/acs.bioconjchem.8b0008612https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXislykt70%253D&md5=29f3a6473705c3549232a73aeba3f385Rapid, Stoichiometric, Site-Specific Modification of Aldehyde-Containing Proteins Using a Tandem Knoevenagel-Intra Michael Addition ReactionYu, Jian; Shen, Da; Zhang, Hanjie; Yin, ZhengBioconjugate Chemistry (2018), 29 (4), 1016-1020CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)A site-specific modification of aldehyde-contg. proteins using a tandem Knoevenagel-intra Michael addn. reaction was developed. The reaction featured fast kinetics (50 M-1s-1) and favorable stoichiometry. Various functionalities could be introduced into the protein with little impact on its function and conformation. The reaction was successfully applied in the labeling of living cells.
- 13An, P.; Lewandowski, T. M.; Erbay, T. G.; Liu, P.; Lin, Q. Sterically Shielded, Stabilized Nitrile Imine for Rapid Bioorthogonal Protein Labeling in Live Cells. J. Am. Chem. Soc. 2018, 140, 4860– 4868, DOI: 10.1021/jacs.8b0012613https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXls12rsrs%253D&md5=06ffc1977b8a9c476be809a06944438cSterically Shielded, Stabilized Nitrile Imine for Rapid Bioorthogonal Protein Labeling in Live CellsAn, Peng; Lewandowski, Tracey M.; Erbay, Tugce G.; Liu, Peng; Lin, QingJournal of the American Chemical Society (2018), 140 (14), 4860-4868CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)In pursuit of fast bioorthogonal reactions, reactive moieties have been increasingly employed for selective labeling of biomols. in living systems, posing a challenge in attaining reactivity without sacrificing selectivity. To address this challenge, here the authors report a bioinspired strategy in which mol. shape controls the selectivity of a transient, highly reactive nitrile imine dipole. By tuning the shape of structural pendants attached to the ortho position of the N-aryl ring of diaryltetrazoles, precursors of nitrile imines, the authors discovered a sterically shielded nitrile imine that favors the 1,3-dipolar cycloaddn. over the competing nucleophilic addn. The photogenerated nitrile imine exhibits an extraordinarily long half-life of 102 s in aq. medium, owing to its unique mol. shape that hinders the approach of a nucleophile as shown by DFT calcns. The utility of this sterically shielded nitrile imine in rapid (∼1 min) bioorthogonal labeling of glucagon receptor in live mammalian cells was demonstrated.
- 14Matsuo, K.; Nishikawa, Y.; Masuda, M.; Hamachi, I. Live-Cell Protein Sulfonylation Based on Proximity-driven N-Sulfonyl Pyridone Chemistry. Angew. Chem., Int. Ed. 2018, 57, 659– 662, DOI: 10.1002/anie.20170797214https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhvFyjsrvN&md5=03016c07f05e88b2836d9746d8e20804Live-Cell Protein Sulfonylation Based on Proximity-driven N-Sulfonyl Pyridone ChemistryMatsuo, Kazuya; Nishikawa, Yuki; Masuda, Marie; Hamachi, ItaruAngewandte Chemie, International Edition (2018), 57 (3), 659-662CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)The development of bioorthogonal approaches for labeling of endogenous proteins under the multimol. crowding conditions of live cells is highly desirable for the anal. and engineering of proteins without using genetic manipulation. N-Sulfonyl pyridone (SP) is reported as a new reactive group for protein sulfonylation. The ligand-directed SP chem. was able to modify not only purified proteins in vitro, but also endogenous ones on the surface of and inside live cells selectively and rapidly, which allowed to convert endogenous proteins to FRET-based biosensors in situ.
- 15Fang, Y. Z.; Zhang, H.; Huang, Z.; Scinto, S. L.; Yang, J. C.; Ende, C. W. A.; Dmitrenko, O.; Johnson, D. S.; Fox, J. M. Photochemical syntheses, transformations, and bioorthogonal chemistry of trans-cycloheptene and sila trans-cycloheptene Ag(I) complexes. Chem. Sci. 2018, 9, 1953– 1963, DOI: 10.1039/C7SC04773H15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXksFOnsg%253D%253D&md5=0ae7c0fd5e5dc85411d62b7b4be3453aPhotochemical syntheses, transformations, and bioorthogonal chemistry of trans-cycloheptene and sila trans-cycloheptene Ag(I) complexesFang, Yinzhi; Zhang, Han; Huang, Zhen; Scinto, Samuel L.; Yang, Jeffrey C.; am Ende, Christopher W.; Dmitrenko, Olga; Johnson, Douglas S.; Fox, Joseph M.Chemical Science (2018), 9 (7), 1953-1963CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)A photochem. synthesis of AgNO3 complexes of trans-cycloheptene (TCH) and trans-1-sila-4-cycloheptene (Si-TCH) derivs. is described. A low temp. flow photoreactor was designed to enable the synthesis of carbocyclic TCH derivs. due to their thermal sensitivity in the absence of metal coordination. Unlike the free carbocycles, TCH•AgNO3 complexes can be handled at rt and stored for weeks in the freezer (-18 °C). Si-TCH•AgNO3 complexes are esp. robust, and are bench stable for days at rt, and for months in the freezer. X-ray crystallog. was used to characterize a Si-TCH•AgNO3 complex for the first time. With decomplexation of AgNO3 in situ, metal-free TCO and Si-TCH derivs. can engage in a range of cycloaddn. reactions as well as dihydroxylation reactions. Computation was used to predict that Si-TCH would engage in bioorthogonal reactions that are more rapid than the most reactive trans-cyclooctenes. Metal-free Si-TCH derivs. were shown to display good stability in soln., and to engage in the fastest bioorthogonal reaction reported to date (k2 1.14 x 107 M-1s-1 in 9:1 H2O:MeOH). Utility in bioorthogonal protein labeling in live cells is described, including labeling of GFP with an unnatural tetrazine-contg. amino acid. The reactivity and specificity of the Si-TCH reagents with tetrazines in live mammalian cells was also evaluated using the HaloTag platform. The cell labeling expts. show that Si-TCH derivs. are best suited as probe mols. in the cellular environment.
- 16Blackman, M. L.; Royzen, M.; Fox, J. M. Tetrazine ligation: fast bioconjugation based on inverse-electron-demand Diels-Alder reactivity. J. Am. Chem. Soc. 2008, 130, 13518– 13519, DOI: 10.1021/ja805380516https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtFChtrjN&md5=9596082bfdbadaea3cb280a633c34db1Tetrazine Ligation: Fast Bioconjugation Based on Inverse-Electron-Demand Diels-Alder ReactivityBlackman, Melissa L.; Royzen, Maksim; Fox, Joseph M.Journal of the American Chemical Society (2008), 130 (41), 13518-13519CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Described is a bioorthogonal reaction that proceeds with unusually fast reaction rates without need for catalysis: the cycloaddn. of s-tetrazine and trans-cyclooctene derivs. The reactions tolerate a broad range of functionality and proceed in high yield in org. solvents, water, cell media, or cell lysate. The rate of the ligation between trans-cyclooctene and 3,6-di-(2-pyridyl)-s-tetrazine is very rapid (k2 2000 M-1 s-1). This fast reactivity enables protein modification at low concn.
- 17Devaraj, N. K.; Upadhyay, R.; Haun, J. B.; Hilderbrand, S. A.; Weissleder, R. Fast and sensitive pretargeted labeling of cancer cells through a tetrazine/trans-cyclooctene cycloaddition. Angew. Chem., Int. Ed. 2009, 48, 7013– 7016, DOI: 10.1002/anie.20090323317https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtV2ltr3P&md5=0caa98e20c8fdd4cf6fec674453b1081Fast and Sensitive Pretargeted Labeling of Cancer Cells through a Tetrazine/trans-Cyclooctene CycloadditionDevaraj, Neal K.; Upadhyay, Rabi; Haun, Jered B.; Hilderbrand, Scott A.; Weissleder, RalphAngewandte Chemie, International Edition (2009), 48 (38), 7013-7016, S7013/1-S7013/6CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)The novel asym. tetrazine I was conjugated to a com. available far-red indocyanine fluorophore, Vivo-Tag 680. This fluorescent tetrazine deriv. was used to image trans-cyclooctene-modified affinity ligands on live cancer cells through a bioorthogonal cycloaddn. with a reaction rate of approx. 6000±200 M-1 s-1 in serum at 37 °C. To maximize the fluorescence signal, up to six trans-cyclooctene moieties were attached to the antibody used to pretarget cells for labeling. Com. available cetuximab was labeled with trans-cyclooctene succinimidyl carbonate and used for pretargeting expts.
- 18Agard, N. J.; Baskin, J. M.; Prescher, J. A.; Lo, A.; Bertozzi, C. R. A comparative study of bioorthogonal reactions with azides. ACS Chem. Biol. 2006, 1, 644– 648, DOI: 10.1021/cb600322818https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtFyhtbfJ&md5=fdee8107c9514d2cc3569d4d5b3df256A comparative study of bioorthogonal reactions with azidesAgard, Nicholas J.; Baskin, Jeremy M.; Prescher, Jennifer A.; Lo, Anderson; Bertozzi, Carolyn R.ACS Chemical Biology (2006), 1 (10), 644-648CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Detection of metabolites and post-translational modifications can be achieved using the azide as a bioorthogonal chem. reporter. Once introduced into target biomols., either metabolically or through chem. modification, the azide can be tagged with probes using one of three highly selective reactions: the Staudinger ligation, the Cu(I)-catalyzed azide-alkyne cycloaddn., or the strain-promoted [3+2] cycloaddn. Here, the authors compared these chemistries in the context of various biol. applications, including labeling of biomols. in complex lysates and on live cell surfaces. The Cu(I)-catalyzed reaction was most efficient for detecting azides in protein samples but was not compatible with live cells due to the toxicity of the reagents. Both the Staudinger ligation and the strain-promoted [3+2] cycloaddn. using optimized cyclooctynes were effective for tagging azides on live cells. The best reagent for this application was dependent upon the specific structure of the azide. These results provide a guide for biologists in choosing a suitable ligation chem.
- 19Dommerholt, J.; van Rooijen, O.; Borrmann, A.; Guerra, C. F.; Bickelhaupt, F. M.; van Delft, F. L. Highly accelerated inverse electron-demand cycloaddition of electron-deficient azides with aliphatic cyclooctynes. Nat. Commun. 2014, 5, 5378, DOI: 10.1038/ncomms637819https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvFansb7N&md5=d0038fe3424275c33a5ebfc561b2bce3Highly accelerated inverse electron-demand cycloaddition of electron-deficient azides with aliphatic cyclooctynesDommerholt, Jan; van Rooijen, Olivia; Borrmann, Annika; Guerra, Celia Fonseca; Bickelhaupt, F. Matthias; van Delft, Floris L.Nature Communications (2014), 5 (), 5378CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)Strain-promoted azide-alkyne cycloaddn. (SPAAC) as a conjugation tool has found broad application in material sciences, chem. biol. and even in vivo use. However, despite tremendous effort, SPAAC remains fairly slow (0.2-0.5 M-1 s-1) and efforts to increase reaction rates by tailoring of cyclooctyne structure have suffered from a poor trade-off between cyclooctyne reactivity and stability. We here wish to report tremendous acceleration of strain-promoted cycloaddn. of an aliph. cyclooctyne (bicyclo[6.1.0]non-4-yne, BCN) with electron-deficient aryl azides, with reaction rate consts. reaching 2.0-2.9 M-1 s-1. A remarkable difference in rate consts. of aliph. cyclooctynes vs. benzoannulated cyclooctynes is noted, enabling a next level of orthogonality by a judicious choice of azide-cyclooctyne combinations, which is inter alia applied in one-pot three-component protein labeling. The pivotal role of azide electronegativity is explained by d.-functional theory calcns. and electronic-structure analyses, which indicates an inverse electron-demand mechanism is operative with an aliph. cyclooctyne.
- 20Kamber, D. N.; Liang, Y.; Blizzard, R. J.; Liu, F.; Mehl, R. A.; Houk, K. N.; Prescher, J. A. 1,2,4-Triazines Are Versatile Bioorthogonal Reagents. J. Am. Chem. Soc. 2015, 137, 8388– 8391, DOI: 10.1021/jacs.5b0510020https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtVansLnM&md5=43ead4d9e9086de4df148decd38e27121,2,4-Triazines Are Versatile Bioorthogonal ReagentsKamber, David N.; Liang, Yong; Blizzard, Robert J.; Liu, Fang; Mehl, Ryan A.; Houk, K. N.; Prescher, Jennifer A.Journal of the American Chemical Society (2015), 137 (26), 8388-8391CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)A new class of bioorthogonal reagents, 1,2,4-triazines, is described. These scaffolds are stable in biol. media and capable of robust reactivity with trans-cyclooctene (TCO). The enhanced stability of the triazine scaffold enabled its direct use in recombinant protein prodn. The triazine-TCO reaction can also be used in tandem with other bioorthogonal cycloaddn. reactions. These features fill current voids in the bioorthogonal toolkit.
- 21Hoffmann, J. E.; Plass, T.; Nikic, I.; Aramburu, I. V.; Koehler, C.; Gillandt, H.; Lemke, E. A.; Schultz, C. Highly Stable trans-Cyclooctene Amino Acids for Live-Cell Labeling. Chem. - Eur. J. 2015, 21, 12266– 12270, DOI: 10.1002/chem.20150164721https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFOrtbjN&md5=632174adf34f4a3986213dadd02479fcHighly Stable trans-Cyclooctene Amino Acids for Live-Cell LabelingHoffmann, Jan-Erik; Plass, Tilman; Nikic, Ivana; Valle Aramburu, Iker; Koehler, Christine; Gillandt, Hartmut; Lemke, Edward A.; Schultz, CarstenChemistry - A European Journal (2015), 21 (35), 12266-12270CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)Trans-Cyclooctene groups incorporated into proteins via non-canonical amino acids (ncAAs) are emerging as specific handles for bioorthogonal chem. Here, we present a highly improved synthetic access to the axially and the equatorially linked trans-cyclooct-2-ene isomers (1 a,b). We further show that the axially connected isomer has a half-life about 10 times higher than the equatorial isomer and reacts with tetrazines much faster, as detd. by stopped-flow expts. The improved properties resulted in different labeling performance of the insulin receptor on the surface of intact cells.
- 22Rossin, R.; van den Bosch, S. M.; Ten Hoeve, W.; Carvelli, M.; Versteegen, R. M.; Lub, J.; Robillard, M. S. Highly reactive trans-cyclooctene tags with improved stability for Diels-Alder chemistry in living systems. Bioconjugate Chem. 2013, 24, 1210– 1217, DOI: 10.1021/bc400153y22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXosFygtrY%253D&md5=04e742d34442b4d0e8704a9be92d1e5fHighly Reactive trans-Cyclooctene Tags with Improved Stability for Diels-Alder Chemistry in Living SystemsRossin, Raffaella; van den Bosch, Sandra M.; ten Hoeve, Wolter; Carvelli, Marco; Versteegen, Ron M.; Lub, Johan; Robillard, Marc S.Bioconjugate Chemistry (2013), 24 (7), 1210-1217CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)One of the challenges of pretargeted radioimmunotherapy, which centers on the capture of a radiolabeled probe by a preinjected tumor-bound antibody, is the potential immunogenicity of biol. capturing systems. A bioorthogonal chem. approach may circumvent this drawback, but effective in vivo chem. in mice, larger animals, and eventually humans, requires very high reagent reactivity, sufficient stability, and retained selectivity. We report here that the reactivity of the fastest bioorthogonal reaction, the inverse-electron-demand-Diels-Alder cycloaddn. between a tetrazine probe and a trans-cyclooctene-tagged antibody, can be increased 10-fold (k2 = 2.7 × 105 M-1s-1) via the trans-cyclooctene, approaching the speed of biol. interactions, while also increasing its stability. This was enabled by the finding that the trans-cyclooctene tag is probably deactivated through isomerization to the unreactive cis-cyclooctene isomer by interactions with copper-contg. proteins, and that increasing the steric hindrance on the tag can impede this process. Next, we found that the higher reactivity of axial vs. equatorial linked TCO can be augmented by the choice of linker. The new, stabilized, and more reactive tag allowed for improved tumor-to-nontumor ratios in pretargeted tumor-bearing mice.
- 23Sletten, E. M.; Bertozzi, C. R. From mechanism to mouse: a tale of two bioorthogonal reactions. Acc. Chem. Res. 2011, 44, 666– 676, DOI: 10.1021/ar200148z23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtVWhurnF&md5=75f5134a99b940e43822840294bc9a5dFrom Mechanism to Mouse: A Tale of Two Bioorthogonal ReactionsSletten, Ellen M.; Bertozzi, Carolyn R.Accounts of Chemical Research (2011), 44 (9), 666-676CODEN: ACHRE4; ISSN:0001-4842. (American Chemical Society)A review. Bioorthogonal reactions are chem. reactions that neither interact with nor interfere with a biol. system. The participating functional groups must be inert to biol. moieties, must selectively reactive with each other under biocompatible conditions, and, for in vivo applications, must be nontoxic to cells and organisms. Addnl., it is helpful if one reactive group is small and therefore minimally perturbing of a biomol. into which it has been introduced either chem. or biosynthetically. Examples from the past decade suggest that a promising strategy for bioorthogonal reaction development begins with an anal. of functional group and reactivity space outside those defined by Nature. Issues such as stability of reactants and products (particularly in water), kinetics, and unwanted side reactivity with biofunctionalities must be addressed, ideally guided by detailed mechanistic studies. Finally, the reaction must be tested in a variety of environments, escalating from aq. media to biomol. solns. to cultured cells and, for the most optimized transformations, to live organisms. Work in our lab. led to the development of two bioorthogonal transformations that exploit the azide as a small, abiotic, and bioinert reaction partner: the Staudinger ligation and strain-promoted azide-alkyne cycloaddn. The Staudinger ligation is based on the classic Staudinger redn. of azides with triarylphosphines first reported in 1919. In the ligation reaction, the intermediate aza-ylide undergoes intramol. reaction with an ester, forming an amide bond faster than aza-ylide hydrolysis would otherwise occur in water. The Staudinger ligation is highly selective and reliably forms its product in environs as demanding as live mice. However, the Staudinger ligation has some liabilities, such as the propensity of phosphine reagents to undergo air oxidn. and the relatively slow kinetics of the reaction. The Staudinger ligation takes advantage of the electrophilicity of the azide; however, the azide can also participate in cycloaddn. reactions. In 1961, Wittig and Krebs noted that the strained, cyclic alkyne cyclooctyne reacts violently when combined neat with Ph azide, forming a triazole product by 1,3-dipolar cycloaddn. This observation stood in stark contrast to the slow kinetics assocd. with 1,3-dipolar cycloaddn. of azides with unstrained, linear alkynes, the conventional Huisgen process. Notably, the reaction of azides with terminal alkynes can be accelerated dramatically by copper catalysis (this highly popular Cu-catalyzed azide-alkyne cycloaddn. (CuAAC) is a quintessential "click" reaction). However, the copper catalysts are too cytotoxic for long-term exposure with live cells or organisms. Thus, for applications of bioorthogonal chem. in living systems, we built upon Wittig and Krebs' observation with the design of cyclooctyne reagents that react rapidly and selectively with biomol.-assocd. azides. This strain-promoted azide-alkyne cycloaddn. is often referred to as "Cu-free click chem.". Mechanistic and theor. studies inspired the design of a series of cyclooctyne compds. bearing fluorine substituents, fused rings, and judiciously situated heteroatoms, with the goals of optimizing azide cycloaddn. kinetics, stability, soly., and pharmacokinetic properties. Cyclooctyne reagents have now been used for labeling azide-modified biomols. on cultured cells and in live Caenorhabditis elegans, zebrafish, and mice. As this special issue testifies, the field of bioorthogonal chem. is firmly established as a challenging frontier of reaction methodol. and an important new instrument for biol. discovery. The above reactions, as well as several newcomers with bioorthogonal attributes, have enabled the high-precision chem. modification of biomols. in vitro, as well as real-time visualization of mols. and processes in cells and live organisms. The consequence is an impressive body of new knowledge and technol., amassed using a relatively small bioorthogonal reaction compendium. Expansion of this toolkit, an effort that is already well underway, is an important objective for chemists and biologists alike.
- 24Link, A. J.; Vink, M. K.; Tirrell, D. A. Presentation and detection of azide functionality in bacterial cell surface proteins. J. Am. Chem. Soc. 2004, 126, 10598– 10602, DOI: 10.1021/ja047629c24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXmtFOktL8%253D&md5=6bbc1f4e362bbe16dfd95b4620ef38abPresentation and detection of azide functionality in bacterial cell surface proteinsLink, A. James; Vink, Mandy K. S.; Tirrell, David A.Journal of the American Chemical Society (2004), 126 (34), 10598-10602CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)An improved protocol for copper-catalyzed triazole formation on the bacterial cell surface is described. Addn. of highly pure CuBr to cells treated with azidohomoalanine (2) leads to ∼. 10-Fold more extensive cell surface labeling than previously obsd. This highly active catalyst allows detection of the methionine analogs azidoalanine (1), azidonorvaline (3), and azidonorleucine (4) in cell surface proteins. Azidoalanine was previously believed to be silent with regard to the cellular protein synthesis machinery.
- 25Tian, F.; Tsao, M. L.; Schultz, P. G. A phage display system with unnatural amino acids. J. Am. Chem. Soc. 2004, 126, 15962– 15963, DOI: 10.1021/ja045673m25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXpslyksLk%253D&md5=32e9325687097a9bb9640da36ae8047cA Phage Display System with Unnatural Amino AcidsTian, Feng; Tsao, Meng-Lin; Schultz, Peter G.Journal of the American Chemical Society (2004), 126 (49), 15962-15963CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)We report a general method to display peptide-contg. unnatural amino acids on filamentous M13 phage. Five distinct unnatural amino acids were site-specifically incorporated at the N-terminal of the M13 phage minor coat protein pIII. Phages that contain p-azidophenylalanine can undergo a highly specific azide-alkyne [3+2] cycloaddn. reaction with an alkyne-derivatized fluorophore. The generalization of phage display to include unnatural amino acids should significantly increase the scope of phage display technol.
- 26Yang, J.; Seckute, J.; Cole, C. M.; Devaraj, N. K. Live-cell imaging of cyclopropene tags with fluorogenic tetrazine cycloadditions. Angew. Chem., Int. Ed. 2012, 51, 7476– 7479, DOI: 10.1002/anie.20120212226https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xos1egs74%253D&md5=c60edddde8754df584b0205368be1e5cLive-Cell Imaging of Cyclopropene Tags with Fluorogenic Tetrazine CycloadditionsYang, Jun; Seckute, Jolita; Cole, Christian M.; Devaraj, Neal K.Angewandte Chemie, International Edition (2012), 51 (30), 7476-7479, S7476/1-S7476/26CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)The authors demonstrated the applicability of methylcyclopropene tags as dienophiles for reaction with fluorogenic tetrazines. Through systematic synthetic modifications, the stability, size, and reactivity of the cyclopropene scaffold were optimized. Methylcyclopropene derivs. were developed that react rapidly with tetrazines while retaining their aq. stability and small size. These cyclopropene handles elicited fluorescent responses from quenched tetrazine dyes and were suitable for cellular imaging applications, which was demonstrated by imaging cyclopropene phospholipids distributed in live human breast cancer cells.
- 27Patterson, D. M.; Nazarova, L. A.; Xie, B.; Kamber, D. N.; Prescher, J. A. Functionalized cyclopropenes as bioorthogonal chemical reporters. J. Am. Chem. Soc. 2012, 134, 18638– 18643, DOI: 10.1021/ja306043627https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsFWjurvL&md5=68598cbedefd059aaa5b63325d7d1ea6Functionalized Cyclopropenes as Bioorthogonal Chemical ReportersPatterson, David M.; Nazarova, Lidia A.; Xie, Bryan; Kamber, David N.; Prescher, Jennifer A.Journal of the American Chemical Society (2012), 134 (45), 18638-18643CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Chem. reporters are unique functional groups that can be used to label biomols. in living systems. Only a handful of broadly applicable reporters have been identified to date, owing to the rigorous demands placed on these functional groups in biol. settings. We describe here a new chem. reporter-cyclopropene-that can be used to target biomols. in vitro and in live cells. A variety of substituted cyclopropene scaffolds were synthesized and found to be stable in aq. soln. and in the presence of biol. nucleophiles. Furthermore, some of the cyclopropene units were metabolically introduced into cell surface glycans and subsequently detected with covalent probes. The small size and selective reactivity of cyclopropenes will facilitate efforts to tag diverse collections of biomols. in vivo.
- 28Andersen, K. A.; Aronoff, M. R.; McGrath, N. A.; Raines, R. T. Diazo groups endure metabolism and enable chemoselectivity in cellulo. J. Am. Chem. Soc. 2015, 137, 2412– 2415, DOI: 10.1021/ja509581528https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXitlWmsrs%253D&md5=de8c0a2a4bd272e46e60057cee01c3bdDiazo Groups Endure Metabolism and Enable Chemoselectivity in CelluloAndersen, Kristen A.; Aronoff, Matthew R.; McGrath, Nicholas A.; Raines, Ronald T.Journal of the American Chemical Society (2015), 137 (7), 2412-2415CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The authors introduce a stabilized diazo group as a reporter for chem. biol. ManDiaz, which is a diazo deriv. of N-acetylmannosamine, is found to endure cellular metab. and label the surface of a mammalian cell. There its diazo group can undergo a 1,3-dipolar cycloaddn. with a strained alkyne, providing a signal comparable to that from the azido congener, ManNAz. The chemoselectivity of diazo and alkynyl groups enables dual labeling of cells that is not possible with azido and alkynyl groups. Thus, the diazo group, which is approx. half the size of an azido group, provides unique opportunities for orthogonal labeling of cellular components.
- 29Karver, M. R.; Weissleder, R.; Hilderbrand, S. A. Bioorthogonal reaction pairs enable simultaneous, selective, multi-target imaging. Angew. Chem., Int. Ed. 2012, 51, 920– 922, DOI: 10.1002/anie.20110438929https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs1SqurfM&md5=d8e69da77db3f495bc3422bd051dfd77Bioorthogonal Reaction Pairs Enable Simultaneous, Selective, Multi-Target ImagingKarver, Mark R.; Weissleder, Ralph; Hilderbrand, Scott A.Angewandte Chemie, International Edition (2012), 51 (4), 920-922, S920/1-S920/12CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)The authors describe the testing and in cell validation of a tetrazine-trans-cyclooctene (TCO) reaction pair that is orthogonal to azide-cyclooctyne cycloaddn. chem. The chosen pairs were able to react concurrently in the same culture to fluorescently label two different cancer cell types. The authors' method uses only small mol. based reagents that also can be readily incorporated into a wide variety of systems and potentially may be used in conjunction with fluorogenic azide and tetrazine substrates to achieve addnl. improvements in specificity and sensitivity. The ability to perform multiple, rapid, simultaneous chem. reactions with a high degree of specificity in chem. complex environments should prove to be a useful tool in chem., biol., and medicine.
- 30Cole, C. M.; Yang, J.; Seckute, J.; Devaraj, N. K. Fluorescent live-cell imaging of metabolically incorporated unnatural cyclopropene-mannosamine derivatives. ChemBioChem 2013, 14, 205– 208, DOI: 10.1002/cbic.20120071930https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXjsleltg%253D%253D&md5=10f4ee328411eac99eab0797301994acFluorescent Live-Cell Imaging of Metabolically Incorporated Unnatural Cyclopropene-Mannosamine DerivativesCole, Christian M.; Yang, Jun; Seckute, Jolita; Devaraj, Neal K.ChemBioChem (2013), 14 (2), 205-208CODEN: CBCHFX; ISSN:1439-4227. (Wiley-VCH Verlag GmbH & Co. KGaA)This study reported that unnatural cyclo-propene-mannosamine derivs. can be used to image glycans on live human cancer cell lines. The results showed that recently developed methylcyclopropene bioorthogonal handles can be utilized for metabolic imaging of unnatural mannosamine derivs. on live-cell surfaces. In general, the data exhibit the possible use of cyclopropenes as reactive mini-tags for a myriad of applications in the profiling and imaging of small mols.
- 31Spate, A. K.; Busskamp, H.; Niederwieser, A.; Schart, V. F.; Marx, A.; Wittmann, V. Rapid labeling of metabolically engineered cell-surface glycoconjugates with a carbamate-linked cyclopropene reporter. Bioconjugate Chem. 2014, 25, 147– 154, DOI: 10.1021/bc400448731https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2c3lvVCqsg%253D%253D&md5=32429d08597db5715b49a065da7df2a1Rapid labeling of metabolically engineered cell-surface glycoconjugates with a carbamate-linked cyclopropene reporterSpate Anne-Katrin; Busskamp Holger; Niederwieser Andrea; Schart Verena F; Marx Andreas; Wittmann ValentinBioconjugate chemistry (2014), 25 (1), 147-54 ISSN:.Metabolic oligosaccharide engineering is a valuable tool to monitor cellular carbohydrates. Here, we report the synthesis of a novel N-acyl-mannosamine derivative bearing a methylcyclopropene tag that is attached to the sugar via a carbamate moiety. This derivative undergoes rapid Diels-Alder reaction with inverse electron demand. We demonstrate that the cell's biosynthetic machinery incorporates this non-natural mannosamine derivative into glycoconjugates that can, subsequently, be labeled within less than 10 min with a new sulfo-Cy3-tetrazine conjugate. Using this tetrazine-dye conjugate for the detection of the methylcyclopropene-tagged mannosamine derivative, we could achieve dual labeling of two different metabolically incorporated sugars combining a Diels-Alder reaction with inverse electron demand and a strain-promoted azide-alkyne cycloaddition which are carried out simultaneously in a single step.
- 32Sachdeva, A.; Wang, K.; Elliott, T.; Chin, J. W. Concerted, rapid, quantitative, and site-specific dual labeling of proteins. J. Am. Chem. Soc. 2014, 136, 7785– 7788, DOI: 10.1021/ja412978932https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXos1Wlurg%253D&md5=58e3a3c448deb78bf2a2e88ed57a8f9aConcerted, Rapid, Quantitative, and Site-Specific Dual Labeling of ProteinsSachdeva, Amit; Wang, Kaihang; Elliott, Thomas; Chin, Jason W.Journal of the American Chemical Society (2014), 136 (22), 7785-7788CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Rapid, one-pot, concerted, site-specific labeling of proteins at genetically encoded unnatural amino acids with distinct small mols. at physiol. pH, temp., and pressure is an important challenge. Current approaches require sequential labeling, low pH, and typically days to reach completion, limiting their utility. We report the efficient, genetically encoded incorporation of alkyne- and cyclopropene-contg. amino acids at distinct sites in a protein using an optimized orthogonal translation system in E. coli. and quant., site-specific, one-pot, concerted protein labeling with fluorophores bearing azide and tetrazine groups, resp. Protein double labeling in aq. buffer at physiol. pH, temp., and pressure is quant. in 30 min.
- 33Rideout, D. Self-assembling cytotoxins. Science 1986, 233, 561– 563, DOI: 10.1126/science.352375733https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL28Xlt12isbc%253D&md5=3abf3eb1b14176e35c5fd91193d5e3bdSelf-assembling cytotoxinsRideout, DarrylScience (Washington, DC, United States) (1986), 233 (4763), 561-3CODEN: SCIEAS; ISSN:0036-8075.Decanal [112-31-2] and N-amino-N'-1-octylguanidine (AOG) [66002-80-0], combined at 28 μM each, mediated erythrocyte lysis within 80 min under physiol. conditions. By contrast, no lysis was obsd. after 20 h with decanal (56 μM) or AOG (100 μM) alone. The pronounced synergism obsd. for these chems. and similar reactive pairs of chems. is due to the self-assembly of more cytotoxic hydrazones in situ. Decanal and AOG also exhibit synergistic activity against cultured human cells (HeLa) and bacteria (Escherichia coli J96). This synergism may be useful in the design of cytotoxins that would self-assemble selectively from nontoxic precursors within tumors, while sparing normal tissue.
- 34Murray, C. W.; Rees, D. C. The rise of fragment-based drug discovery. Nat. Chem. 2009, 1, 187– 192, DOI: 10.1038/nchem.21734https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXmtF2rs78%253D&md5=0bd1a40fd927cce9200f0fa296a7462fThe rise of fragment-based drug discoveryMurray, Christopher W.; Rees, David C.Nature Chemistry (2009), 1 (3), 187-192CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)A review. The search for new drugs is plagued by high attrition rates at all stages in research and development. Chemists have an opportunity to tackle this problem because attrition can be traced back, in part, to the quality of the chem. leads. Fragment-based drug discovery (FBDD) is a new approach, increasingly used in the pharmaceutical industry, for reducing attrition and providing leads for previously intractable biol. targets. FBDD identifies low-mol.-wt. ligands (∼150 Da) that bind to biol. important macromols. The three-dimensional exptl. binding mode of these fragments is detd. using X-ray crystallog. or NMR spectroscopy, and is used to facilitate their optimization into potent mols. with drug-like properties. Compared with high-throughput-screening, the fragment approach requires fewer compds. to be screened, and, despite the lower initial potency of the screening hits, offers more efficient and fruitful optimization campaigns. Here, we review the rise of FBDD, including its application to discovering clin. candidates against targets for which other chem. approaches have struggled.
- 35Thirumurugan, P.; Matosiuk, D.; Jozwiak, K. Click chemistry for drug development and diverse chemical-biology applications. Chem. Rev. 2013, 113, 4905– 4979, DOI: 10.1021/cr200409f35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXkslahtLY%253D&md5=d9c664c199b63c7d556c60fe9cd1507dClick Chemistry for Drug Development and Diverse Chemical-Biology ApplicationsThirumurugan, Prakasam; Matosiuk, Dariusz; Jozwiak, KrzysztofChemical Reviews (Washington, DC, United States) (2013), 113 (7), 4905-4979CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review with 303 refs. on application of lick chem. reactions in chem., biochem., and pharmacol.
- 36Lewis, W. G.; Green, L. G.; Grynszpan, F.; Radic, Z.; Carlier, P. R.; Taylor, P.; Finn, M. G.; Sharpless, K. B. Click chemistry in situ: acetylcholinesterase as a reaction vessel for the selective assembly of a femtomolar inhibitor from an array of building blocks. Angew. Chem., Int. Ed. 2002, 41, 1053– 1057, DOI: 10.1002/1521-3773(20020315)41:6<1053::AID-ANIE1053>3.0.CO;2-436https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XisVeisbo%253D&md5=e1ff6aedcbf7ae1523f4e3082e801c86Click chemistry in situ: Acetylcholinesterase as a reaction vessel for the selective assembly of a femtomolar inhibitor from an array of building blocksLewis, Warren G.; Green, Luke G.; Grynszpan, Flavio; Radic, Zoran; Carlier, Paul R.; Taylor, Palmer; Finn, M. G.; Sharpless, K. BarryAngewandte Chemie, International Edition (2002), 41 (6), 1053-1057CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH)Form-fitting chem. in a protein mold is enabled by the use of the 1,3-dipolar cycloaddn. of azides and alkynes. The enzyme acetylcholinesterase preferentially assembles one pair of these reactants, each of which bears a group that binds to adjacent positions on the protein structure, into a 1,2,3-triazole adduct that is the most potent noncovalent inhibitor of the enzyme yet developed.
- 37Mocharla, V. P.; Colasson, B.; Lee, L. V.; Roper, S.; Sharpless, K. B.; Wong, C. H.; Kolb, H. C. In situ click chemistry: enzyme-generated inhibitors of carbonic anhydrase II. Angew. Chem., Int. Ed. 2005, 44, 116– 120, DOI: 10.1002/anie.20046158037https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXptFaj&md5=9b890f3eecd0ede6132a45c90aff9607In situ click chemistry: Enzyme-generated inhibitors of carbonic anhydrase IIMocharla, Vani P.; Colasson, Benoit; Lee, Lac V.; Roeper, Stefanie; Sharpless, K. Barry; Wong, Chi-Huey; Kolb, Hartmuth C.Angewandte Chemie, International Edition (2005), 44 (1), 116-120CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Target-guided synthesis: Rather than making and screening thousands of compds. for lead discovery, in situ click chem. employs the biol. target itself to assemble its inhibitors by selectively binding and interconnecting reagents within the confines of its binding sites. Subnanomolar inhibitors of carbonic anhydrase II were produced by the enzyme from simple azide and acetylene precursors.
- 38Whiting, M.; Muldoon, J.; Lin, Y. C.; Silverman, S. M.; Lindstrom, W.; Olson, A. J.; Kolb, H. C.; Finn, M. G.; Sharpless, K. B.; Elder, J. H.; Fokin, V. V. Inhibitors of HIV-1 protease by using in situ click chemistry. Angew. Chem., Int. Ed. 2006, 45, 1435– 1439, DOI: 10.1002/anie.20050216138https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XitF2qtLg%253D&md5=d575780546dbc314eb9df85bc2459539Inhibitors of HIV-1 protease by using in situ click chemistryWhiting, Matthew; Muldoon, John; Lin, Ying-Chuan; Silverman, Steven M.; Lindstrom, William; Olson, Arthur J.; Kolb, Hartmuth C.; Finn, M. G.; Sharpless, K. Barry; Elder, John H.; Fokin, Valery V.Angewandte Chemie, International Edition (2006), 45 (9), 1435-1439CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Twice poor equals potent: HIV-1 Protease assembles its own potent inhibitor through formation of the triazole linkage from azide- and alkyne-contg. fragments that are themselves poor binders.
- 39Devaraj, N. K.; Weissleder, R. Biomedical applications of tetrazine cycloadditions. Acc. Chem. Res. 2011, 44, 816– 827, DOI: 10.1021/ar200037t39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXmslOnu74%253D&md5=7c8ef732eea8dbf9e11b8bd4e4ddeef0Biomedical Applications of Tetrazine CycloadditionsDevaraj, Neal K.; Weissleder, RalphAccounts of Chemical Research (2011), 44 (9), 816-827CODEN: ACHRE4; ISSN:0001-4842. (American Chemical Society)A review. Disease mechanisms are increasingly being resolved at the mol. level. Biomedical success at this scale creates synthetic opportunities for combining specifically designed orthogonal reactions in applications such as imaging, diagnostics, and therapy. For practical reasons, it would be helpful if bioorthogonal coupling reactions proceeded with extremely rapid kinetics (k > 103 M-1 s-1) and high specificity. Improving kinetics would minimize both the time and amt. of labeling agent required to maintain high coupling yields. In this Account, we discuss our recent efforts to design extremely rapid bioorthogonal coupling reactions between tetrazines and strained alkenes. These selective reactions were first used to covalently couple conjugated tetrazine near-IR-emitting fluorophores to dienophile-modified extracellular proteins on living cancer cells. Confocal fluorescence microscopy demonstrated efficient and selective labeling, and control expts. showed minimal background fluorescence. Multistep techniques were optimized to work with nanomolar concns. of labeling agent over a time scale of minutes: the result was successful real-time imaging of covalent modification. We subsequently discovered fluorogenic probes that increase in fluorescence intensity after the chem. reaction, leading to an improved signal-to-background ratio. Fluorogenic probes were used for intracellular imaging of dienophiles. We further developed strategies to react and image chemotherapeutics, such as trans-cyclooctene taxol analogs, inside living cells. Because the coupling partners are small mols. (<300 Da), they offer unique steric advantages in multistep amplification. We also describe recent success in using tetrazine reactions to label biomarkers on cells with magneto-fluorescent nanoparticles. Two-step protocols that use bioorthogonal chem. can significantly amplify signals over both one-step labeling procedures as well as two-step procedures that use more sterically hindered biotin-avidin interactions. Nanoparticles can be detected with fluorescence or magnetic resonance techniques. These strategies are now being routinely used on clin. samples for biomarker profiling to predict malignancy and patient outcome. Finally, the authors discuss recent results with tetrazine reactions used for in vivo mol. imaging applications. Rapid tetrazine cycloaddns. allow modular labeling of small mols. with the most commonly used positron emission tomog. isotope, 18F. Addnl., recent work has applied this reaction directly in vivo for the pretargeted imaging of solid tumors. Future work with tetrazine cycloaddns. will undoubtedly lead to optimized protocols, improved probes, and addnl. biomedical applications.
- 40Lebraud, H.; Wright, D. J.; Johnson, C. N.; Heightman, T. D. Protein Degradation by In-Cell Self-Assembly of Proteolysis Targeting Chimeras. ACS Cent. Sci. 2016, 2, 927– 934, DOI: 10.1021/acscentsci.6b0028040https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvFOltbbJ&md5=4cc9b4aaba6ee03cd2d4ea97a72728a5Protein Degradation by In-Cell Self-Assembly of Proteolysis Targeting ChimerasLebraud, Honorine; Wright, David J.; Johnson, Christopher N.; Heightman, Tom D.ACS Central Science (2016), 2 (12), 927-934CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)Selective degrdn. of proteins by proteolysis targeting chimeras (PROTACs) offers a promising potential alternative to protein inhibition for therapeutic intervention. Current PROTAC mols. incorporate a ligand for the target protein, a linker, and an E3 ubiquitin ligase recruiting group, which bring together target protein and ubiquitinating machinery. Such hetero-bifunctional mols. require significant linker optimization and possess high mol. wt., which can limit cellular permeation, soly., and other drug-like properties. We show here that the hetero-bifunctional mol. can be formed intracellularly by bio-orthogonal click combination of two smaller precursors. We designed a tetrazine tagged thalidomide deriv. which reacts rapidly with a trans-cyclo-octene tagged ligand of the target protein in cells to form a cereblon E3 ligase recruiting PROTAC mol. The in-cell click-formed proteolysis targeting chimeras (CLIPTACs) were successfully used to degrade two key oncol. targets, BRD4 and ERK1/2. ERK1/2 degrdn. was achieved using a CLIPTAC based on a covalent inhibitor. We expect this approach to be readily extendable to other inhibitor-protein systems because the tagged E3 ligase recruiter is capable of undergoing the click reaction with a suitably tagged ligand of any protein of interest to elicit its degrdn.
- 41Versteegen, R. M.; Rossin, R.; ten Hoeve, W.; Janssen, H. M.; Robillard, M. S. Click to release: instantaneous doxorubicin elimination upon tetrazine ligation. Angew. Chem., Int. Ed. 2013, 52, 14112– 14116, DOI: 10.1002/anie.20130596941https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvVClsb%252FM&md5=c926aeb4b257768962ca1b3d6285a0aaClick to Release: Instantaneous Doxorubicin Elimination upon Tetrazine LigationVersteegen, Ron M.; Rossin, Raffaella; ten Hoeve, Wolter; Janssen, Henk M.; Robillard, Marc S.Angewandte Chemie, International Edition (2013), 52 (52), 14112-14116CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)We have developed a new bioorthogonal elimination reaction that enables instantaneous, self-immolative, and traceless release of doxorubicin from trans-cyclooctene following tetrazine ligation. In this proof-of-principle study, the inverse-electron-demand Diels-Alder (inv-DA) pyridazine elimination reached 79 % yield within minutes under ambient conditions at micromolar concns.
- 42Xu, M.; Tu, J.; Franzini, R. M. Rapid and efficient tetrazine-induced drug release from highly stable benzonorbornadiene derivatives. Chem. Commun. 2017, 53, 6271– 6274, DOI: 10.1039/C7CC03477F42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXosVymu7s%253D&md5=3b00491f3ba7b323ed3e1a6b84e8950aRapid and efficient tetrazine-induced drug release from highly stable benzonorbornadiene derivativesXu, Minghao; Tu, Julian; Franzini, Raphael M.Chemical Communications (Cambridge, United Kingdom) (2017), 53 (46), 6271-6274CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)A novel class of bioorthogonal release reactions based on benzonorbornadiene derivs. was developed. These carrier mols. are highly stable at physiol. conditions, but react rapidly with 1,2,4,5-tetrazines, and near-quant. release cargo mols. such as drugs and optical reporters.
- 43Neumann, K.; Jain, S.; Gambardella, A.; Walker, S. E.; Valero, E.; Lilienkampf, A.; Bradley, M. Tetrazine-Responsive Self-immolative Linkers. ChemBioChem 2017, 18, 91– 95, DOI: 10.1002/cbic.20160056043https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitVyqsr7I&md5=d677f5477f4872930382cf2e1d3bb12eTetrazine-Responsive Self-immolative LinkersNeumann, Kevin; Jain, Sarthak; Gambardella, Alessia; Walker, Sarah E.; Valero, Elsa; Lilienkampf, Annamaria; Bradley, MarkChemBioChem (2017), 18 (1), 91-95CODEN: CBCHFX; ISSN:1439-4227. (Wiley-VCH Verlag GmbH & Co. KGaA)Mols. that undergo activation or modulation following the addn. of benign external small-mol. chem. stimuli have numerous applications. Here, we report the highly efficient "decaging" of a variety of moieties by activation of a "self-immolative" linker, by application of water-sol. and stable tetrazine, including the controlled delivery of doxorubicin in a cellular context.
- 44Jimenez-Moreno, E.; Guo, Z.; Oliveira, B. L.; Albuquerque, I. S.; Kitowski, A.; Guerreiro, A.; Boutureira, O.; Rodrigues, T.; Jimenez-Oses, G.; Bernardes, G. J. Vinyl Ether/Tetrazine Pair for the Traceless Release of Alcohols in Cells. Angew. Chem., Int. Ed. 2017, 56, 243– 247, DOI: 10.1002/anie.20160960744https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitVGhtrzP&md5=e22f3ae8a0963e02028a8ee180bc7d09Vinyl Ether/Tetrazine Pair for the Traceless Release of Alcohols in CellsJimenez-Moreno, Ester; Guo, Zijian; Oliveira, Bruno L.; Albuquerque, Ines S.; Kitowski, Annabel; Guerreiro, Ana; Boutureira, Omar; Rodrigues, Tiago; Jimenez-Oses, Gonzalo; Bernardes, Goncalo J. L.Angewandte Chemie, International Edition (2017), 56 (1), 243-247CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)The cleavage of a protecting group from a protein or drug under bioorthogonal conditions enables accurate spatiotemporal control over protein or drug activity. Disclosed herein is that vinyl ethers serve as protecting groups for alc.-contg. mols. and as reagents for bioorthogonal bond-cleavage reactions. A vinyl ether moiety was installed in a range of mols., including amino acids, a monosaccharide, a fluorophore, and an analog of the cytotoxic drug duocarmycin. Tetrazine-mediated decaging proceeded under biocompatible conditions with good yields and reasonable kinetics. Importantly, the nontoxic, vinyl ether duocarmycin double prodrug was successfully decaged in live cells to reinstate cytotoxicity. This bioorthogonal reaction presents broad applicability and may be suitable for in vivo applications.
- 45Mejia Oneto, J. M.; Khan, I.; Seebald, L.; Royzen, M. In Vivo Bioorthogonal Chemistry Enables Local Hydrogel and Systemic Pro-Drug To Treat Soft Tissue Sarcoma. ACS Cent. Sci. 2016, 2, 476– 482, DOI: 10.1021/acscentsci.6b0015045https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtFCqs77O&md5=b7a01ddbbf126ed981be5bb0dc2226a8In Vivo Bioorthogonal Chemistry Enables Local Hydrogel and Systemic Pro-Drug To Treat Soft Tissue SarcomaMejia Oneto, Jose M.; Khan, Irfan; Seebald, Leah; Royzen, MaksimACS Central Science (2016), 2 (7), 476-482CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)The ability to activate drugs only at desired locations avoiding systemic immunosuppression and other dose limiting toxicities is highly desirable. Here we present a new approach, named local drug activation, that uses bioorthogonal chem. to conc. and activate systemic small mols. at a location of choice. This method is independent of endogenous cellular or environmental markers and only depends on the presence of a preimplanted biomaterial near a desired site (e.g., tumor). We demonstrate the clear therapeutic benefit with minimal side effects of this approach in mice over systemic therapy using a doxorubicin pro-drug against xenograft tumors of a type of soft tissue sarcoma (HT1080).
- 46Zhang, G.; Li, J.; Xie, R.; Fan, X.; Liu, Y.; Zheng, S.; Ge, Y.; Chen, P. R. Bioorthogonal Chemical Activation of Kinases in Living Systems. ACS Cent. Sci. 2016, 2, 325– 331, DOI: 10.1021/acscentsci.6b0002446https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmslWgsrw%253D&md5=ebb651cbb2fc3dca008863104c9a94d5Bioorthogonal Chemical Activation of Kinases in Living SystemsZhang, Gong; Li, Jie; Xie, Ran; Fan, Xinyuan; Liu, Yanjun; Zheng, Siqi; Ge, Yun; Chen, Peng R.ACS Central Science (2016), 2 (5), 325-331CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)Selective manipulation of protein kinases under living conditions is highly desirable yet extremely challenging, particularly in a gain-of-function fashion. Here we employ our recently developed bioorthogonal cleavage reaction as a general strategy for intracellular activation of individual kinases. Site-specific incorporation of trans-cyclooctene-caged lysine in place of the conserved catalytic lysine, in conjunction with the cleavage partner dimethyl-tetrazine, allowed efficient lysine decaging with the kinase activity chem. rescued in living systems.
- 47Li, J.; Jia, S.; Chen, P. R. Diels-Alder reaction-triggered bioorthogonal protein decaging in living cells. Nat. Chem. Biol. 2014, 10, 1003– 1005, DOI: 10.1038/nchembio.165647https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvVGiurfM&md5=4723cef7d02aec13267b61d08c9517a6Diels-Alder reaction-triggered bioorthogonal protein decaging in living cellsLi, Jie; Jia, Shang; Chen, Peng R.Nature Chemical Biology (2014), 10 (12), 1003-1005CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Small mols. that specifically activate an intracellular protein of interest are highly desirable. A generally applicable strategy, however, remains elusive. Herein we describe a small mol.-triggered bioorthogonal protein decaging technique that relies on the inverse electron-demand Diels-Alder reaction for eliminating a chem. caged protein side chain within living cells. This method permits the efficient activation of a given protein (for example, an enzyme) in its native cellular context within minutes.
- 48Wu, H.; Alexander, S. C.; Jin, S.; Devaraj, N. K. A Bioorthogonal Near-Infrared Fluorogenic Probe for mRNA Detection. J. Am. Chem. Soc. 2016, 138, 11429– 11432, DOI: 10.1021/jacs.6b0162548https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xhtlamt77K&md5=5d18f87589f5d989b9b00655dd8b0663A Bioorthogonal Near-Infrared Fluorogenic Probe for mRNA DetectionWu, Haoxing; Alexander, Seth C.; Jin, Shuaijiang; Devaraj, Neal K.Journal of the American Chemical Society (2016), 138 (36), 11429-11432CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)There is significant interest in developing methods that visualize and detect RNA in live cells. Bioorthogonal template driven tetrazine ligations could be a powerful route to visualizing nucleic acids in native cells, yet past work has been limited with respect to the diversity of fluorogens that can be activated via a tetrazine reaction. Herein we report a novel bioorthogonal tetrazine uncaging reaction that harnesses tetrazine reactivity to unmask vinyl ether caged fluorophores spanning the visible spectrum, including a near-IR (NIR) emitting cyanine dye. Vinyl ether caged fluorophores and tetrazine partners are conjugated to high affinity antisense nucleic acid probes, which show highly selective fluorogenic reactivity when annealed to their resp. target RNA sequences. A target sequence in the 3' untranslated region (3' UTR) of an expressed mRNA was successfully detected in live cells employing appropriate nucleic acid probes bearing a tetrazine reactive NIR fluorogen. Given the expansion of tetrazine fluorogenic chem. to NIR dyes, we believe highly selective proximity induced fluorogenic tetrazine reactions could find broad uses in illuminating endogenous biomols. in living cells.
- 49Ji, X.; Zhou, C.; Ji, K.; Aghoghovbia, R. E.; Pan, Z.; Chittavong, V.; Ke, B.; Wang, B. Click and Release: A Chemical Strategy toward Developing Gasotransmitter Prodrugs by Using an Intramolecular Diels-Alder Reaction. Angew. Chem., Int. Ed. 2016, 55, 15846– 15851, DOI: 10.1002/anie.20160873249https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvFahtb3P&md5=a4888e92153a23269644879744e14a12Click and Release: A Chemical Strategy toward Developing Gasotransmitter Prodrugs by Using an Intramolecular Diels-Alder ReactionJi, Xingyue; Zhou, Cheng; Ji, Kaili; Aghoghovbia, Robert E.; Pan, Zhixiang; Chittavong, Vayou; Ke, Bowen; Wang, BingheAngewandte Chemie, International Edition (2016), 55 (51), 15846-15851CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Prodrug strategies have been proven to be a very effective way of addressing delivery problems. Much of the chem. in prodrug development relies on the ability to mask an appropriate functional group, which can be removed under appropriate conditions. However, developing org. prodrugs of gasotransmitters represent unique challenges. This is esp. true with carbon monoxide, which does not have an easy "handle" for bioreversible derivatization. By taking advantage of an intramol. Diels-Alder reaction, we have developed a prodrug strategy for prepns. of org. CO prodrugs (I, where X = O, NR1 and n = 1,2) that are stable during synthesis and storage, and yet readily release CO with tunable release rates under near physiol. conditions. The effectiveness of the CO prodrug system in delivering a sufficient quantity of CO for possible therapeutic applications has been studied using a cell culture anti-inflammatory assay and a colitis animal model. These studies fully demonstrate the proof of concept, and lay a strong foundation for further medicinal chem. work in developing org. CO prodrugs.
- 50Carlson, J. C. T.; Mikula, H.; Weissleder, R. Unraveling Tetrazine-Triggered Bioorthogonal Elimination Enables Chemical Tools for Ultrafast Release and Universal Cleavage. J. Am. Chem. Soc. 2018, 140, 3603– 3612, DOI: 10.1021/jacs.7b1121750https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitVKrtr8%253D&md5=0330db3cb69894d1d11e2343c98a4da0Unraveling tetrazine-triggered bioorthogonal elimination enables chemical tools for ultrafast release and universal cleavageCarlson, Jonathan C. T.; Mikula, Hannes; Weissleder, RalphJournal of the American Chemical Society (2018), 140 (10), 3603-3612CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Recent developments in bond cleavage reactions have expanded the scope of bioorthogonal chem. beyond click ligation and enabled new strategies for probe activation and therapeutic delivery. These applications, however, remain in their infancy, with further innovations needed to achieve the efficiency required for versatile and broadly useful tools in vivo. Among these chemistries, the tetrazine/trans-cyclooctene click-to-release reaction has exemplary kinetics and adaptability but achieves only partial release and is incompletely understood, which has limited its application. Investigating the mechanistic features of this reaction's performance, we discovered profound pH sensitivity, exploited it with acid-functionalized tetrazines that both enhance and markedly accelerate release, and ultimately uncovered an unexpected dead-end isomer as the reason for poor release. Implementing facile methods to prevent formation of this dead end, we have achieved exceptional efficiency, with essentially complete release across the full scope of physiol. pH, potentiating drug-delivery strategies and expanding the dynamic range of bioorthogonal on/off control.
- 51El-Sagheer, A. H.; Brown, T. Synthesis and polymerase chain reaction amplification of DNA strands containing an unnatural triazole linkage. J. Am. Chem. Soc. 2009, 131, 3958– 3964, DOI: 10.1021/ja806589651https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXisVCjtL0%253D&md5=c64505e86e67b410762714ad9af48364Synthesis and Polymerase Chain Reaction Amplification of DNA Strands Containing an Unnatural Triazole LinkageEl-Sagheer, Afaf H.; Brown, TomJournal of the American Chemical Society (2009), 131 (11), 3958-3964CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)DNA strands contg. an unnatural T-triazole-T linkage have been synthesized by click DNA ligation between oligonucleotides with 3'-AZT and 5'-propargylamido dT and amplified efficiently by polymerase chain reaction (PCR) using several different polymerases. DNA sequencing of PCR amplicons and clones in two different sequence contexts revealed the presence of a single thymidine at the ligation site. The remarkable ability of thermostable polymerases to reproducibly copy DNA templates contg. such an unnatural backbone opens up intriguing possibilities in gene synthesis, genetic anal., biol., and nanotechnol.
- 52Budin, I.; Devaraj, N. K. Membrane assembly driven by a biomimetic coupling reaction. J. Am. Chem. Soc. 2012, 134, 751– 753, DOI: 10.1021/ja207687352https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs12hsL3I&md5=25dbc6361458e7ff63b6cec654587696Membrane Assembly Driven by a Biomimetic Coupling ReactionBudin, Itay; Devaraj, Neal K.Journal of the American Chemical Society (2012), 134 (2), 751-753CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)One of the major goals of synthetic biol. is the development of non-natural cellular systems. In this work, we describe a catalytic biomimetic coupling reaction capable of driving the de novo self-assembly of phospholipid membranes. Our system features a copper-catalyzed azide-alkyne cycloaddn. that results in the formation of a triazole-contg. phospholipid analog. Concomitant assembly of membranes occurs spontaneously, not requiring preexisting membranes to house catalysts or precursors. The substitution of efficient synthetic reactions for key biochem. processes may offer a general route toward synthetic biol. systems.
- 53Cendret, V.; Francois-Heude, M.; Mendez-Ardoy, A.; Moreau, V.; Fernandez, J. M.; Djedaini-Pilard, F. Design and synthesis of a ″click″ high-mannose oligosaccharide mimic emulating Man8 binding affinity towards Con A. Chem. Commun. 2012, 48, 3733– 3735, DOI: 10.1039/c2cc30773aThere is no corresponding record for this reference.
- 54Dawson, P. E.; Muir, T. W.; Clark-Lewis, I.; Kent, S. B. Synthesis of proteins by native chemical ligation. Science 1994, 266, 776– 779, DOI: 10.1126/science.797362954https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXitVGgtrw%253D&md5=1d352d817ab48851500610dcd32d1136Synthesis of proteins by native chemical ligationDawson, Philip E.; Muir, Tom W.; Clark-Lewis, Ian; Kent, Stephen B. H.Science (Washington, D. C.) (1994), 266 (5186), 776-9CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)A simple technique has been devised that allows the direct synthesis of native backbone proteins of moderate size. Chemoselective reaction of two unprotected peptide segments gives an initial thioester-linked species. Spontaneous rearrangement of this transient intermediate yields a full-length product with a native peptide bond at the ligation site. The utility of native chem. ligation was demonstrated by the one-step prepn. of a cytokine contg. multiple disulfides. The polypeptide ligation product was folded and oxidized to form the native disulfide-contg. protein mol. Native chem. ligation is an important step toward the general application of chem. to proteins.
- 55de Figueiredo, R. M.; Suppo, J. S.; Campagne, J. M. Nonclassical Routes for Amide Bond Formation. Chem. Rev. 2016, 116, 12029– 12122, DOI: 10.1021/acs.chemrev.6b0023755https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2svktV2ktQ%253D%253D&md5=833714af5bdc3786efdb953edfae36ecNonclassical Routes for Amide Bond Formationde Figueiredo Renata Marcia; Suppo Jean-Simon; Campagne Jean-MarcChemical reviews (2016), 116 (19), 12029-12122 ISSN:.The present review offers an overview of nonclassical (e.g., with no pre- or in situ activation of a carboxylic acid partner) approaches for the construction of amide bonds. The review aims to comprehensively discuss relevant work, which was mainly done in the field in the last 20 years. Organization of the data follows a subdivision according to substrate classes: catalytic direct formation of amides from carboxylic and amines ( section 2 ); the use of carboxylic acid surrogates ( section 3 ); and the use of amine surrogates ( section 4 ). The ligation strategies (NCL, Staudinger, KAHA, KATs, etc.) that could involve both carboxylic acid and amine surrogates are treated separately in section 5 .
- 56Nilsson, B. L.; Kiessling, L. L.; Raines, R. T. High-yielding Staudinger ligation of a phosphinothioester and azide to form a peptide. Org. Lett. 2001, 3, 9– 12, DOI: 10.1021/ol006739v56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXovVOjurg%253D&md5=fff7cb4d30c89b152983e3e5f50f157dHigh-Yielding Staudinger Ligation of a Phosphinothioester and Azide To Form a PeptideNilsson, Bradley L.; Kiessling, Laura L.; Raines, Ronald T.Organic Letters (2001), 3 (1), 9-12CODEN: ORLEF7; ISSN:1523-7060. (American Chemical Society)The Staudinger ligation can be used to couple a peptide with a C-terminal phosphinothioester to another with an N-terminal α-azido group to form a single peptide that contains no residual atoms of the phosphinothiol entity. Here, diphenylphosphinomethanethiol thioesters are shown to give high isolated yields for this transformation. This finding provides precedent for a powerful and versatile new method for the total synthesis of proteins, avoiding the native chem. ligation method's reliance on the presence of a cysteine residue for bond formation.
- 57Saxon, E.; Armstrong, J. I.; Bertozzi, C. R. A ″traceless″ Staudinger ligation for the chemoselective synthesis of amide bonds. Org. Lett. 2000, 2, 2141– 2143, DOI: 10.1021/ol006054v57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXktVGktrk%253D&md5=88b7c588c348dd4b59b996db08c3d0a1A "Traceless" Staudinger Ligation for the Chemoselective Synthesis of Amide BondsSaxon, Eliana; Armstrong, Joshua I.; Bertozzi, Carolyn R.Organic Letters (2000), 2 (14), 2141-2143CODEN: ORLEF7; ISSN:1523-7060. (American Chemical Society)A novel modification is reported of the authors' previously reported Staudinger ligation that generates an amide bond from an azide and a specifically functionalized phosphine. This method for the selective formation of an amide bond, which does not require the orthogonal protection of distal functional groups, should find general utility in synthetic and biol. chem. 2-AcOC6H4PPh2 and (1-acetyl-2-imidazolyl)diphenylphosphine reacted with an azidonucleoside (5'-azido-N-benzoyl-2',5'-dideoxyadenosine) in wet THF to convert the azido group to an acetylamino group in >95% yield.
- 58Erben, A.; Grossmann, T. N.; Seitz, O. DNA-triggered synthesis and bioactivity of proapoptotic peptides. Angew. Chem., Int. Ed. 2011, 50, 2828– 2832, DOI: 10.1002/anie.20100710358https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXivVGhurs%253D&md5=b4096818c6a8bd987fd1c8e7d913c661DNA-Triggered Synthesis and Bioactivity of Proapoptotic PeptidesErben, Anne; Grossmann, Tom N.; Seitz, OliverAngewandte Chemie, International Edition (2011), 50 (12), 2828-2832, S2828/1-S2828/17CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Diseases are frequently caused by changes in the genetic infrastructure. In such cases, the disordered state of a diseased cell is encoded in the DNA and reflected in the level and sequence of the expressed RNA mols. The information obtained from nucleic acids may be used to direct mol. therapies only to diseased cells and tissues. In a fascinating approach, disease-specific nucleic acid sequences could be hijacked to trigger the formation or release of drug mols. Herein a reaction system is introduced in which the sequence information of an unstructured DNA template is used to trigger the transfer of an aminoacyl group from a donating thioester-modified peptide-nucleic acid (PNA) conjugate to an acceptor peptidyl-PNA conjugate. It is demonstrated that the template can act as a catalyst which instructs the formation of many product mols. per template mol. The formed peptide-PNA conjugate was designed to interfere with the protein-protein interactions between caspase-9, a protease involved in the initiation of programmed cell death (apoptosis), and the X-linked inhibitor of apoptosis protein XIAP. It is shown that the nucleic acid programmed peptide synthesis allows activation of caspase-9 and a downstream caspase.
- 59Noda, H.; Eros, G.; Bode, J. W. Rapid ligations with equimolar reactants in water with the potassium acyltrifluoroborate (KAT) amide formation. J. Am. Chem. Soc. 2014, 136, 5611– 5614, DOI: 10.1021/ja501844259https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXltFGltLc%253D&md5=755cfa1e934bab498e8ac3c9d67d8009Rapid ligations with equimolar reactants in water with the potassium acyltrifluoroborate (KAT) amide formationNoda, Hidetoshi; Eros, Gabor; Bode, Jeffrey W.Journal of the American Chemical Society (2014), 136 (15), 5611-5614CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The identification of fast, chemoselective bond-forming reactions is one of the major contemporary challenges in chem. We show that chemoselective amide-forming ligations of potassium acyltrifluoroborates (KATs) and O-carbamoylhydroxylamines proceed in the presence of all unprotected functional groups with a second-order rate const. of 20 M-1 s-1. PEG chains, lipids, biotin, and dyes were introduced onto an unprotected 31-mer peptide (GLP-1 analog) with equimolar ratios of reactants within minutes at 1 mM and within 1 h at 100 μM, even with Mw 20 000 PEG. This conjugation reaction provides a new approach to the synthesis of mols. such as protein-protein and protein-polymer conjugates.
- 60Rudd, A. K.; Devaraj, N. K. Traceless Synthesis of Ceramides in Living Cells Reveals Saturation-Dependent Apoptotic Effects. Proc. Natl. Acad. Sci. USA , 2018, In Press. DOI: DOI: 10.1073/pnas.1804266115 .There is no corresponding record for this reference.
- 61Mehl, R. A.; Anderson, J. C.; Santoro, S. W.; Wang, L.; Martin, A. B.; King, D. S.; Horn, D. M.; Schultz, P. G. Generation of a bacterium with a 21 amino acid genetic code. J. Am. Chem. Soc. 2003, 125, 935– 939, DOI: 10.1021/ja028415361https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXhtlCmug%253D%253D&md5=fdbba595b21142ebd43fbdaf399008aaGeneration of a Bacterium with a 21 Amino Acid Genetic CodeMehl, Ryan A.; Anderson, J. Christopher; Santoro, Stephen W.; Wang, Lei; Martin, Andrew B.; King, David S.; Horn, David M.; Schultz, Peter G.Journal of the American Chemical Society (2003), 125 (4), 935-939CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)We have generated a completely autonomous bacterium with a 21 amino acid genetic code. This bacterium can biosynthesize a nonstandard amino acid from basic carbon sources and incorporate this amino acid into proteins in response to the amber nonsense codon. The biosynthetic pathway for the amino acid p-aminophenylalanine (pAF) as well as a unique pAF synthetase and cognate tRNA were added to Escherichia coli. Denaturing gel electrophoresis and mass spectrometric anal. show that pAF is incorporated into myoglobin with fidelity and efficiency rivaling those of the common 20 amino acids. This and other such organisms may provide an opportunity to examine the evolutionary consequences of adding new amino acids to the genetic repertoire, as well as generate proteins with new or enhanced biol. functions.
- 62Bohannon, M. B.; Kleiman, R. Cyclopropene fatty acids of selected seed oils from bombacaceae, malvaceae, and sterculiaceae. Lipids 1978, 13, 270– 273, DOI: 10.1007/BF02533669There is no corresponding record for this reference.
- 63Bao, X.; Katz, S.; Pollard, M.; Ohlrogge, J. Carbocyclic fatty acids in plants: biochemical and molecular genetic characterization of cyclopropane fatty acid synthesis of Sterculiafoetida. Proc. Natl. Acad. Sci. U. S. A. 2002, 99, 7172– 7177, DOI: 10.1073/pnas.092152999There is no corresponding record for this reference.
- 64Chen, K.; Huang, X.; Kan, S. B. J.; Zhang, R. K.; Arnold, F. H. Enzymatic construction of highly strained carbocycles. Science 2018, 360, 71– 75, DOI: 10.1126/science.aar423964https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXmvFamsL0%253D&md5=df5bbb212f61f1f29ed867125d53dcdeEnzymatic construction of highly strained carbocyclesChen, Kai; Huang, Xiongyi; Kan, S. B. Jennifer; Zhang, Ruijie K.; Arnold, Frances H.Science (Washington, DC, United States) (2018), 360 (6384), 71-75CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Small carbocycles are structurally rigid and possess high intrinsic energy due to their ring strain. These features lead to broad applications but also create challenges for their construction. We report the engineering of a cytochrome P 450 variant (designated P411) that catalyze the formation of chiral bicyclobutanes, one of the most strained four-membered systems, via successive carbene addn. to unsatd. carbon-carbon bonds. Enzymes that produce cyclopropenes, putative intermediates to the bicyclobutanes, were also identified. These genetically encoded proteins are readily optimized by directed evolution, function in Escherichia coli, and act on structurally diverse substrates with high efficiency and selectivity, providing an effective route to many chiral strained structures. This biotransformation is easily performed at preparative scale, and the resulting strained carbocycles can be derivatized, opening myriad potential applications.
- 65Lappchen, T.; Rossin, R.; van Mourik, T. R.; Gruntz, G.; Hoeben, F. J. M.; Versteegen, R. M.; Janssen, H. M.; Lub, J.; Robillard, M. S. DOTA-tetrazine probes with modified linkers for tumor pretargeting. Nucl. Med. Biol. 2017, 55, 19– 26, DOI: 10.1016/j.nucmedbio.2017.09.00165https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1M7gtlCnug%253D%253D&md5=c8e55fde4872a6407e12fce3812fe901DOTA-tetrazine probes with modified linkers for tumor pretargetingLappchen Tilman; Rossin Raffaella; van Mourik Tiemen R; Gruntz Guillaume; Lub Johan; Hoeben Freek J M; Versteegen Ron M; Janssen Henk M; Robillard Marc SNuclear medicine and biology (2017), 55 (), 19-26 ISSN:.INTRODUCTION: Pretargeted radioimmunoimaging and -therapy approaches building on the bioorthogonal inverse-electron-demand Diels-Alder (IEDDA) reaction between strained trans-cyclooctenes (TCO) and electron-deficient tetrazines (Tz) have yielded impressive results in recent years and have proven a vital alternative to biological pretargeting systems. After improvement of the TCO-antibody conjugates, we here report on our evaluation of a new series of radiolabeled Tz-probes. METHODS: Four new Tz-probes were synthesized, radiolabeled with lutetium-177, and characterized in vitro in terms of lipophilicity, reactivity, and stability in PBS and mouse serum. The in vivo biodistribution profile and tumor-targeting potential of the probes were evaluated in LS174T tumor-bearing mice pretargeted with TCO-antibody conjugates using non-pretargeted mice as control. RESULTS: Radiolabeling of all probes proceeded in high yields providing the (177)Lu-labeled tetrazines in >95% radiochemical purity without any further purification. In mouse serum, half-lives of the probes varied between 8 and 13 h, with the exception of the most lipophilic probe, [(177)Lu]1b, with a serum half-life of less than 1 h. This probe also showed the fastest blood clearance (t1/2 = 5.4 min), more than 2-fold faster than PEG-linked probes [(177)Lu]3 and [(177)Lu]4, and even 3-fold faster than the other small probes without the PEG-linker, [(177)Lu]1a and [(177)Lu]2. In the pretargeting experiments, tumor uptake of the lead probe [(177)Lu]4 (~6 %ID/g) was most closely approached by [(177)Lu]2, followed by [(177)Lu]3 and [(177)Lu]1a. While all the smaller and more lipophilic probes suffered from increased liver uptake, the PEG-linked probe [(177)Lu]3 with its additional negative charge surprisingly showed the highest kidney uptake among all of the probes. CONCLUSION: The in vitro performance of some of the new tetrazine probes turned out to be comparable to the established lead probe [(177)Lu]Lu-DOTA-PEG11-Tz ([(177)Lu]4). However, tumor pretargeting studies in vivo showed lower tumor uptake and increased uptake in non-target organs.
- 66Houghton, J. L.; Membreno, R.; Abdel-Atti, D.; Cunanan, K. M.; Carlin, S.; Scholz, W. W.; Zanzonico, P. B.; Lewis, J. S.; Zeglis, B. M. Establishment of the In Vivo Efficacy of Pretargeted Radioimmunotherapy Utilizing Inverse Electron Demand Diels-Alder Click Chemistry. Mol. Cancer Ther. 2017, 16, 124– 133, DOI: 10.1158/1535-7163.MCT-16-050366https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjsF2hsQ%253D%253D&md5=eb72c9c0f7b38994358445c8586f9570Establishment of the In Vivo Efficacy of Pretargeted Radioimmunotherapy Utilizing Inverse Electron Demand Diels-Alder Click ChemistryHoughton, Jacob L.; Membreno, Rosemery; Abdel-Atti, Dalya; Cunanan, Kristen M.; Carlin, Sean; Scholz, Wolfgang W.; Zanzonico, Pat B.; Lewis, Jason S.; Zeglis, Brian M.Molecular Cancer Therapeutics (2017), 16 (1), 124-133CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)The pretargeting system based on the inverse electron demand Diels-Alder reaction (IEDDA) between trans-cyclooctene (TCO) and tetrazine (Tz) combines the favorable pharmacokinetic properties of radiolabeled small mols. with the affinity and specificity of antibodies. This strategy has proven to be an efficient method for the molecularly targeted delivery of pharmaceuticals, including isotopes for radiol. imaging. Despite encouraging results from in vivo PET imaging studies, this promising system has yet to be thoroughly evaluated for pretargeted radioimmunotherapy (PRIT). Toward that end, we synthesized two novel 177Lu-labeled tetrazine-bearing radioligands. Next, we compared the usefulness of our ligands for PRIT when paired with TCO-modified 5B1-a human, anti-CA19.9 mAb-in preclin. murine models of pancreatic cancer. The exemplary ligand, 177Lu-DOTA-PEG7-Tz, showed rapid (4.6 ± 0.8% ID/g at 4 h) and persistent (16.8 ± 3.9% ID/g at 120 h) uptake in tumors while concurrently clearing from blood and nontarget tissues. Single-dose therapy studies using 5B1-TCO and varying amts. of 177Lu-DOTA-PEG7-Tz (400, 800, and 1,200μCi) showed that our system elicits a dose-dependent therapeutic response in mice bearing human xenografts. Furthermore, dosimetry calcns. suggest that our approach is amenable to clin. applications with its excellent dosimetric profile in organs of clearance (i.e., liver and kidneys) as well as in dose-limiting tissues, such as red marrow. This study established that a pretargeted methodol. utilizing the IEDDA reaction can rapidly and specifically deliver a radiotherapeutic payload to tumor tissue, thus illustrating its excellent potential for clin. translation.
- 67Goodwin, D. A.; Meares, C. F.; McCall, M. J.; McTigue, M.; Chaovapong, W. Pre-targeted immunoscintigraphy of murine tumors with indium-111-labeled bifunctional haptens. J. Nucl. Med. 1988, 29 (2), 226– 234There is no corresponding record for this reference.
- 68Zeglis, B. M.; Sevak, K. K.; Reiner, T.; Mohindra, P.; Carlin, S. D.; Zanzonico, P.; Weissleder, R.; Lewis, J. S. A pretargeted PET imaging strategy based on bioorthogonal Diels-Alder click chemistry. J. Nucl. Med. 2013, 54, 1389– 1396, DOI: 10.2967/jnumed.112.11584068https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVaqu7fF&md5=a930e7e2f9b22e030b692399ff53baafA pretargeted PET imaging strategy based on bioorthogonal Diels-Alder click chemistryZeglis, Brian M.; Sevak, Kuntal K.; Reiner, Thomas; Mohindra, Priya; Carlin, Sean D.; Zanzonico, Pat; Weissleder, Ralph; Lewis, Jason S.Journal of Nuclear Medicine (2013), 54 (8), 1389-1396CODEN: JNMEAQ; ISSN:0161-5505. (Society of Nuclear Medicine and Molecular Imaging)The specificity of antibodies have made immunoconjugates promising vectors for the delivery of radioisotopes to cancer cells; however, their long pharmacol. half-lives necessitate the use of radioisotopes with long phys. half-lives, a combination that leads to high radiation doses to patients. Therefore, the development of targeting modalities that harness the advantages of antibodies without their pharmacokinetic limitations is desirable. To this end, we report the development of a methodol. for pretargeted PET imaging based on the bioorthogonal Diels-Alder click reaction between tetrazine and transcyclooctene. Methods: A proof-of-concept system based on the A33 antibody, SW1222 colorectal cancer cells, and 64Cu was used. The huA33 antibody was covalently modified with transcyclooctene, and a NOTA-modified tetrazine was synthesized and radiolabeled with 64Cu. Pretargeted in vivo biodistribution and PET imaging expts. were performed with athymic nude mice bearing A33 antigen-expressing, SW1222 colorectal cancer xenografts. Results: The huA33 antibody was modified with transcyclooctene to produce a conjugate with high immunoreactivity, and the 64Cu-NOTA-labeled tetrazine ligand was synthesized with greater than 99% purity and a specific activity of 9-10 MBq/μg. For in vivo expts., mice bearing SW1222 xenografts were injected with transcyclooctene-modified A33; after allowing 24 h for accumulation of the antibody in the tumor, the mice were injected with 64Cu-NOTA-labeled tetrazine for PET imaging and biodistribution expts. At 12 h after injection, the retention of uptake in the tumor (4.1 ± 0.3 percent injected dose per g), coupled with the fecal excretion of excess radioligand, produced images with high tumor-to-background ratios. PET imaging and biodistribution expts. performed using A33 directly labeled with either 64Cu or 89Zr revealed that although abs. tumor uptake was higher with the directly radiolabeled antibodies, the pretargeted system yielded comparable images and tumor-to-muscle ratios at 12 and 24 h after injection. Further, dosimetry calcns. revealed that the 64Cu pretargeting system resulted in only a fraction of the absorbed background dose of A33 directly labeled with 89Zr (0.0124 mSv/MBq vs. 0.4162 mSv/MBq, resp.). Conclusion: The high quality of the images produced by this pretargeting approach, combined with the ability of the methodol. to dramatically reduce nontarget radiation doses to patients, marks this system as a strong candidate for clin. translation.
- 69Rossin, R.; Verkerk, P. R.; van den Bosch, S. M.; Vulders, R. C.; Verel, I.; Lub, J.; Robillard, M. S. In vivo chemistry for pretargeted tumor imaging in live mice. Angew. Chem., Int. Ed. 2010, 49, 3375– 3378, DOI: 10.1002/anie.20090629469https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXlt1Glsbo%253D&md5=5b08f7ad878d9f823dbdd6b4096b71e1In Vivo Chemistry for Pretargeted Tumor Imaging in Live MiceRossin, Raffaella; Renart Verkerk, Pascal; van den Bosch, Sandra M.; Vulders, Roland C. M.; Verel, Iris; Lub, Johan; Robillard, Marc S.Angewandte Chemie, International Edition (2010), 49 (19), 3375-3378, S3375/1-S3375/22CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Bioorthogonal chem. reaction between two exogenous moieties in living animal was used for cancer imaging.
- 70Devaraj, N. K.; Weissleder, R.; Hilderbrand, S. A. Tetrazine-based cycloadditions: application to pretargeted live cell imaging. Bioconjugate Chem. 2008, 19, 2297– 2299, DOI: 10.1021/bc800444670https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsVWrtbfJ&md5=f68878b74748e037b97e9a1f37178250Tetrazine-Based Cycloadditions: Application to Pretargeted Live Cell ImagingDevaraj, Neal K.; Weissleder, Ralph; Hilderbrand, Scott A.Bioconjugate Chemistry (2008), 19 (12), 2297-2299CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Bioorthogonal tetrazine cycloaddns. have been applied to live cell labeling. Tetrazines react irreversibly with the strained dienophile norbornene forming dihydropyrazine products and dinitrogen. The reaction is high yielding, selective, and fast in aq. media. Her2/neu receptors on live human breast cancer cells were targeted with a monoclonal antibody modified with a norbornene. Tetrazines conjugated to a near-IR fluorochrome selectively and rapidly label the pretargeted antibody in the presence of serum. These findings indicate that this chem. is suitable for in vitro labeling expts., and suggests that it may prove a useful strategy for in vivo pretargeted imaging under numerous modalities.