Inducible, Site-Specific Protein Labeling by Tyrosine Oxidation–Strain-Promoted (4 + 2) CycloadditionClick to copy article linkArticle link copied!
- Jorick J. Bruins
- Adrie H. Westphal
- Bauke Albada
- Koen Wagner
- Lina Bartels
- Hergen Spits
- Willem J. H. van Berkel
- Floris L. van Delft
Abstract
Genetically encoded tyrosine (Y-tag) can be utilized as a latent anchor for inducible and site-selective conjugation. Upon oxidation of tyrosine with mushroom tyrosinase, strain-promoted cycloaddition (SPOCQ) of the resulting 1,2-quinone with various bicyclo[6.1.0]nonyne (BCN) derivatives led to efficient conjugation. The method was applied for fluorophore labeling of laminarinase A and for the site-specific preparation of an antibody–drug conjugate.
Introduction
Results and Discussion
Laminarinase A
Figure 1
Figure 1. SPOCQ labeling of G4Y-tagged laminarinase A by reaction of BCN-modified reagent 1 with in situ generated 1,2-quinone. Typical reaction conditions: LamA (1.0 mg/mL), mTyr (0.3 mg/mL), and 1 (4 equiv) in 50 mM potassium phosphate buffer pH 7.3, containing 135 mM NaCl and 10% DMSO as cosolvent.
Figure 2
Figure 2. (A) SDS-PAGE analysis of SPOCQ on LamA–G4Y and wt-LamA. (B) MS profile of LamA–G4Y. (C) MS profile of LamA–G4Y after SPOCQ with 1.
Trastuzumab
AT1002
Figure 3
Figure 3. (A) Schematic representation of G4Y-tagged antibodies. (B) SDS-PAGE analysis of SPOCQ on AT1002[LC]G4Y and wt-AT1002. (C) MS profile of AT1002[LC]G4Y (light chain only). (D) MS spectrum of AT1002[LC]G4Y after SPOCQ with 1. (E) MS profile of AT1002[LC]G4Y after SPOCQ with 2.
Conclusions
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.bioconjchem.7b00046.
Additional method and material details, SPOCQ and expression details, gene and protein sequences, a schematic view of the reaction and corresponding mass values, and MS data. (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgment
Remon van Geel and Peter van Galen are acknowledged for providing kind assistance with the mass spectrometry measurements. This work is funded by the NWO Gravity Program Institute for Chemical Immunology (ICI).
References
This article references 32 other publications.
- 1Foley, T. L. and Burkart, M. D. (2007) Site-specific protein modification: advances and applications Curr. Opin. Chem. Biol. 11, 12– 19 DOI: 10.1016/j.cbpa.2006.11.036Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhsFOhs70%253D&md5=b527edf8f4a8be8dbe10b4f36aa87937Site-specific protein modification: Advances and applicationsFoley, Timothy L.; Burkart, Michael D.Current Opinion in Chemical Biology (2007), 11 (1), 12-19CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)A review. Although chem. methods to modify proteins in a sequence-specific manner have yet to be developed, site-specific post-translational modification of proteins has recently emerged as a major focus in biol. chem. Post-translational modification with functionalized substrate analogs opens up several unique avenues to induce selective reactivity into proteins in a sequence-specific manner, and can be applied to protein identification and manipulation in both in vitro and in vivo contexts. Further in vivo applications of this method will enable the imaging of cellular processes, avoiding nonspecific labeling and probe scattering, major complications obsd. in nonenzymic methods. Addnl., new tools for in vitro protein modification have been developed that offer more versatile ways to study protein structure and function.
- 2Jung, S. and Kwon, I. (2016) Expansion of bioorthogonal chemistries towards site-specific polymer-protein conjugation Polym. Chem. 7, 4584– 98 DOI: 10.1039/C6PY00856AGoogle Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XpvFeisrw%253D&md5=d8ee4ce063caa8dd73b0ab48da2780b4Expansion of bioorthogonal chemistries towards site-specific polymer-protein conjugationJung, Secheon; Kwon, InchanPolymer Chemistry (2016), 7 (28), 4584-4598CODEN: PCOHC2; ISSN:1759-9962. (Royal Society of Chemistry)Polymer conjugation to proteins has been widely used to improve or expand protein properties. However, polymer conjugation to random sites of a target protein often led to a significant loss of crit. protein properties. In order to overcome this, polymer conjugation to specific sites of a protein was developed using the site-specific introduction of non-natural amino acids and bioorthogonal chemistries. This review summarizes the recent advances in bioorthogonal chemistries. As the repertoire of bioorthogonal chemistries available for polymer conjugation is expanding, the site-specific polymer conjugation technique would be a valuable platform technique to design novel protein-polymer conjugates.
- 3Spicer, C. D. and Davis, B. G. (2014) Selective chemical protein modification Nat. Commun. 5, 4740 DOI: 10.1038/ncomms5740Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXksVeksb4%253D&md5=055c882ff0405b0bbb91edca3bcef8cbSelective chemical protein modificationSpicer, Christopher D.; Davis, Benjamin G.Nature Communications (2014), 5 (), 4740CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)The chem. modification of proteins is an important tool for probing natural systems and synthesizing novel conjugates. Here, Spicer and Davis review the merits and limitations of the most useful methods for selective modification at both natural and unnatural amino acids.
- 4Zhang, Z., Smith, B. A., Wang, L., Brock, A., Cho, C., and Schultz, P. G. (2003) A new strategy for the site-specific modification of proteins in vivo Biochemistry 42, 6735– 46 DOI: 10.1021/bi0300231Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXjs12qtr8%253D&md5=07c3108f4ce3e4a107c45f6f70db9bb1A New Strategy for the Site-Specific Modification of Proteins in VivoZhang, Zhiwen; Smith, Brian A. C.; Wang, Lei; Brock, Ansgar; Cho, Charles; Schultz, Peter G.Biochemistry (2003), 42 (22), 6735-6746CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)We recently developed a method for genetically incorporating unnatural amino acids site-specifically into proteins expressed in Escherichia coli in response to the amber nonsense codon. Here we describe the selection of an orthogonal tRNA-TyrRS pair that selectively and efficiently incorporates m-acetyl-L-phenylalanine into proteins in E. coli. We demonstrate that proteins contg. m-acetyl-L-phenylalanine or p-acetyl-L-phenylalanine can be selectively labeled with hydrazide derivs. not only in vitro but also in living cells. The labeling reactions are selective and in general proceed with yields of >75%. In specific examples, m-acetyl-L-phenylalanine was substituted for Lys7 of the cytoplasmic protein Z domain, and for Arg200 of the outer membrane protein LamB, and the mutant proteins were selectively labeled with a series of fluorescent dyes. The genetic incorporation of a nonproteinogenic "ketone handle" into proteins provides a powerful tool for the introduction of biophys. probes for the structural and functional anal. of proteins in vitro or in vivo.
- 5Umeda, A., Thibodeaux, G. N., Zhu, J., Lee, Y., and Zhang, Z. J. (2009) Site-specific protein cross-linking with genetically incorporated 3,4-dihydroxy-L-phenylalanine ChemBioChem 10, 1302– 04 DOI: 10.1002/cbic.200900127Google ScholarThere is no corresponding record for this reference.
- 6Burdine, L., Gillette, T. G., Lin, H. J., and Kodadek, T. (2004) Periodate-triggered cross-linking of DOPA-containing peptide-protein complexes J. Am. Chem. Soc. 126, 11442– 3 DOI: 10.1021/ja045982cGoogle ScholarThere is no corresponding record for this reference.
- 7Mao, H., Hart, S. A., Schink, A., and Pollok, B. A. (2004) Sortase-mediated protein ligation: a new method for protein engineering J. Am. Chem. Soc. 126, 2670– 1 DOI: 10.1021/ja039915eGoogle Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhtVGisrs%253D&md5=7ae77a5a866ce5e2544550754f086560Sortase-Mediated Protein Ligation: A New Method for Protein EngineeringMao, Hongyuan; Hart, Scott A.; Schink, Amy; Pollok, Brian A.Journal of the American Chemical Society (2004), 126 (9), 2670-2671CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Sortase (SrtA), a transpeptidase from Staphylococcus aureus, catalyzes a cell-wall sorting reaction at an LPXTG motif by cleaving between threonine and glycine and subsequently joining the carboxyl group of threonine to an amino group of pentaglycine on the cell wall peptidoglycan. We have applied this transpeptidyl activity of sortase to in vitro protein ligation. We found that in the presence of sortase, protein/peptide with an LPXTG motif can be specifically ligated to an aminoglycine protein/peptide via an amide bond. Addnl., sortase can even conjugate substrates such as (D)-peptides, synthetic branched peptides, and aminoglycine-derivatized small mols. to the C terminus of a recombinant protein. The sortase-mediate protein ligation is robust, specific, and easy to perform, and can be widely applied to specific protein conjugation with polypeptides or mols. of unique biochem. and biophys. properties.
- 8Schumacher, D., Helma, J., Mann, F. A., Pichler, G., Natale, F., Krause, E., Cardoso, M. C., Hackenberger, C. P. R., and Leonhardt, H. (2015) Versatile and efficient site-specific protein functionalization by tubulin tyrosine ligase Angew. Chem., Int. Ed. 54, 13787– 91 DOI: 10.1002/anie.201505456Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFGkt7%252FL&md5=5801b9d620c4656b8ce79e6d96d06e72Versatile and Efficient Site-Specific Protein Functionalization by Tubulin Tyrosine LigaseSchumacher, Dominik; Helma, Jonas; Mann, Florian A.; Pichler, Garwin; Natale, Francesco; Krause, Eberhard; Cardoso, M. Cristina; Hackenberger, Christian P. R.; Leonhardt, HeinrichAngewandte Chemie, International Edition (2015), 54 (46), 13787-13791CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)A novel chemoenzymic approach for simple and fast site-specific protein labeling is reported. Recombinant tubulin tyrosine ligase (TTL) was repurposed to attach various unnatural tyrosine derivs. as small bioorthogonal handles to proteins contg. a short tubulin-derived recognition sequence (Tub-tag). This novel strategy enables a broad range of high-yielding and fast chemoselective C-terminal protein modifications on isolated proteins or in cell lysates for applications in biochem., cell biol., and beyond, as demonstrated by the site-specific labeling of nanobodies, GFP, and ubiquitin.
- 9Holder, P. G., Jones, L. C., Drake, P. M., Barfield, R. M., Banas, S., de Hart, G. W., Baker, J., and Rabuka, D. (2015) Reconstitution of formylglycine-generating enzyme with copper(II) for aldehyde tag conversion J. Biol. Chem. 290, 15730– 45 DOI: 10.1074/jbc.M115.652669Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtVKhsLvN&md5=580235fe160698373c76b21ce354d61bReconstitution of Formylglycine-generating Enzyme with Copper(II) for Aldehyde Tag ConversionHolder, Patrick G.; Jones, Lesley C.; Drake, Penelope M.; Barfield, Robyn M.; Banas, Stefanie; de Hart, Gregory W.; Baker, Jeanne; Rabuka, DavidJournal of Biological Chemistry (2015), 290 (25), 15730-15745CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)To further our aim of synthesizing aldehyde-tagged proteins for research and biotechnol. applications, we developed methods for recombinant prodn. of aerobic formylglycine-generating enzyme (FGE) in good yield. We then optimized the FGE biocatalytic reaction conditions for conversion of cysteine to formylglycine in aldehyde tags on intact monoclonal antibodies. During the development of these conditions, we discovered that pretreating FGE with copper(II) is required for high turnover rates and yields. After further investigation, we confirmed that both aerobic prokaryotic (Streptomyces coelicolor) and eukaryotic (Homo sapiens) FGEs contain a copper cofactor. The complete kinetic parameters for both forms of FGE are described, along with a proposed mechanism for FGE catalysis that accounts for the copper-dependent activity.
- 10Zhang, H., Trout, W. S., Liu, S., Andrade, G. A., Hudson, D. A., Scinto, S. L., Dicker, K. T., Li, Y., Lazouski, N., and Rosenthal, J. 2016, Rapid Bioorthogonal Chemistry Turn-on through Enzymatic or Long Wavelength Photocatalytic Activation of Tetrazine Ligation J. Am. Chem. Soc. 138, 5978– 5983 DOI: 10.1021/jacs.6b02168Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmtFSntrw%253D&md5=701ccea88f61e6eaf883d240e1785833Rapid Bioorthogonal Chemistry Turn-on through Enzymatic or Long Wavelength Photocatalytic Activation of Tetrazine LigationZhang, Han; Trout, William S.; Liu, Shuang; Andrade, Gabriel A.; Hudson, Devin A.; Scinto, Samuel L.; Dicker, Kevin T.; Li, Yi; Lazouski, Nikifar; Rosenthal, Joel; Thorpe, Colin; Jia, Xinqiao; Fox, Joseph M.Journal of the American Chemical Society (2016), 138 (18), 5978-5983CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Rapid bioorthogonal reactivity can be induced by controllable, catalytic stimuli using air as the oxidant. Methylene blue (4 μM) irradiated with red light (660 nm) catalyzes the rapid oxidn. of a dihydrotetrazine to a tetrazine thereby turning on reactivity toward trans-cyclooctene dienophiles. Alternately, the aerial oxidn. of dihydrotetrazines can be efficiently catalyzed by nanomolar levels of horseradish peroxidase under peroxide-free conditions. Selection of dihydrotetrazine/tetrazine pairs of sufficient kinetic stability in aerobic aq. solns. is key to the success of these approaches. In this work, polymer fibers carrying latent dihydrotetrazines were catalytically activated and covalently modified by trans-cyclooctene conjugates of small mols., peptides, and proteins. In addn. to visualization with fluorophores, fibers conjugated to a cell adhesive peptide exhibited a dramatically increased ability to mediate contact guidance of cells.
- 11McGaughey, G. B., Gagne, M., and Rappe, A. K. (1998) pi-Stacking interactions. Alive and well in proteins J. Biol. Chem. 273, 15458– 15463 DOI: 10.1074/jbc.273.25.15458Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXktVSmtLw%253D&md5=ffa442b0a21dd4df1199900542428332π-stacking interactions. Alive and well in proteinsMcgaughey, Georgia B.; Gagne, Marc; Rappe, Anthony K.Journal of Biological Chemistry (1998), 273 (25), 15458-15463CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)A representative set of high resoln. x-ray crystal structures of nonhomologous proteins was examd. to det. the preferred positions and orientations of noncovalent interactions between the arom. side-chains of the amino acids, Phe, Tyr, His, and Trp. To study the primary interactions between arom. amino acids, care was taken to examine only isolated pairs (dimers) of amino acids because trimers and higher order clusters of arom. amino acids behave differently than their dimer counterparts. It was found that pairs (dimers) of arom. side-chain amino acids preferentially aligned their resp. arom. rings in an off-centered parallel orientation. Further, it was found that this parallel-displaced structure was 0.5-0.75 kcal/mol more stable than a T-shaped structure for Phe interactions and 1 kcal/mol more stable than a T-shaped structure for the full set of arom. side-chain amino acids. This exptl. detd. structure and energy difference was consistent with ab initio and mol. mechanics calcns. of the benzene dimer; however, the results were not in agreement with previously published analyses of arom. amino acids in proteins. The preferred orientation was referred to as parallel displaced π-stacking.
- 12Struck, A. W., Bennett, M. R., Shepherd, S. A., Law, B. J., Zhuo, Y., Wong, L. S., and Micklefield, J. (2016) An enzyme cascade for selective modification of tyrosine residues in structurally diverse peptides and proteins J. Am. Chem. Soc. 138, 3038– 45 DOI: 10.1021/jacs.5b10928Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XisVWmu7g%253D&md5=cc3ae0bb185682b8f997b844b0e2dbbcAn Enzyme Cascade for Selective Modification of Tyrosine Residues in Structurally Diverse Peptides and ProteinsStruck, Anna-Winona; Bennett, Matthew R.; Shepherd, Sarah A.; Law, Brian J. C.; Zhuo, Ying; Wong, Lu Shin; Micklefield, JasonJournal of the American Chemical Society (2016), 138 (9), 3038-3045CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Bioorthogonal chem. enables a specific moiety in a complex biomol. to be selectively modified in the presence of many reactive functional groups and other cellular entities. Such selectivity has become indispensable in biol., enabling biomols. to be derivatized, conjugated, labeled, or immobilized for imaging, biochem. assays, or therapeutic applications. Methyltransferase enzymes (MTase) that accept analogs of the cofactor S-adenosyl methionine have been widely deployed for alkyl-diversification and bioorthogonal labeling. However, MTases typically possess tight substrate specificity. Here we introduce a more flexible methodol. for selective derivatization of phenolic moieties in complex biomols. Our approach relies on the tandem enzymic reaction of a fungal tyrosinase and the mammalian catechol-O-methyltransferase (COMT), which can effect the sequential hydroxylation of the phenolic group to give an intermediate catechol moiety that is subsequently O-alkylated. When used in this combination, the alkoxylation is highly selective for tyrosine residues in peptides and proteins, yet remarkably tolerant to changes in the peptide sequence. Tyrosinase-COMT are shown to provide highly versatile and regioselective modification of a diverse range of substrates including peptide antitumor agents, hormones, cyclic peptide antibiotics, and model proteins.
- 13Schlick, T. L., Ding, Z., Kovacs, E. W., and Francis, M. B. (2005) Dual-surface modification of the tobacco mosaic virus J. Am. Chem. Soc. 127, 3718– 23 DOI: 10.1021/ja046239nGoogle Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhs1ant7w%253D&md5=417dfb81edc43b89ef121f8c67ed653aDual-surface modification of the Tobacco mosaic virusSchlick, Tara L.; Ding, Zhebo; Kovacs, Ernest W.; Francis, Matthew B.Journal of the American Chemical Society (2005), 127 (11), 3718-3723CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The protein shell of the tobacco mosaic virus (TMV) provides a robust and practical tubelike scaffold for the prepn. of nanoscale materials. To expand the range of applications for which the capsid can be used, two synthetic strategies have been developed for the attachment of new functionality to either the exterior or the interior surface of the virus. The first of these is accomplished using a highly efficient diazonium coupling/oxime formation sequence, which installs >2000 copies of a material component on the capsid exterior. Alternatively, the inner cavity of the tube can be modified by attaching amines to glutamic acid side chains through a carbodiimide coupling reaction. Both of these reactions have been demonstrated for a series of substrates, including biotin, chromophores, and crown ethers. Through the attachment of PEG polymers to the capsid exterior, org.-sol. TMV rods have been prepd. Finally, the orthogonality of these reactions has been demonstrated by installing different functional groups on the exterior and interior surfaces of the same capsid assemblies.
- 14Ban, H., Gavrilyuk, J., and Barbas, C. F., 3rd (2010) Tyrosine bioconjugation through aqueous ene-type reactions: a click-like reaction for tyrosine J. Am. Chem. Soc. 132, 1523– 5 DOI: 10.1021/ja909062qGoogle Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXkt1Wrug%253D%253D&md5=876ee34e149ceed4f70f7fd4efbb25baTyrosine bioconjugation through aqueous ene-type reactions: A click-like reaction for tyrosineBan, Hitoshi; Gavrilyuk, Julia; Barbas, Carlos F.Journal of the American Chemical Society (2010), 132 (5), 1523-1525CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)A new and versatile class of cyclic diazodicarboxamides that reacts efficiently and selectively with phenols and the phenolic side chain of tyrosine through an ene-like reaction is reported. This mild aq. tyrosine ligation reaction works over a broad pH range and expands the repertoire of aq. chemistries available for small mol., peptide, and protein modification. The tyrosine ligation reactions are shown to be compatible with the labeling of native enzymes and antibodies in a buffered aq. soln. This reaction provides a novel synthetic approach to bispecific antibodies. The authors believe this reaction will find broad utility in peptide and protein chem. and in the chem. of phenol-contg. compds.
- 15Ban, H., Nagano, M., Gavrilyuk, J., Hakamata, W., Inokuma, T., and Barbas, C. F., 3rd (2013) Facile and stabile linkages through tyrosine: bioconjugation strategies with the tyrosine-click reaction Bioconjugate Chem. 24, 520– 32 DOI: 10.1021/bc300665tGoogle Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXkslWlsrY%253D&md5=7f548b32c1e16dd72adbe15b05bbf38cFacile and Stabile Linkages through Tyrosine: Bioconjugation Strategies with the Tyrosine-Click ReactionBan, Hitoshi; Nagano, Masanobu; Gavrilyuk, Julia; Hakamata, Wataru; Inokuma, Tsubasa; Barbas, Carlos F., IIIBioconjugate Chemistry (2013), 24 (4), 520-532CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)The scope, chemoselectivity, and utility of the click-like tyrosine labeling reaction with 4-phenyl-3H-1,2,4-triazoline-3,5(4H)-diones (PTADs) is reported. To study the utility and chemoselectivity of PTAD derivs. in peptide and protein chem., we synthesized PTAD derivs. possessing azide, alkyne, and ketone groups and studied their reactions with amino acid derivs. and peptides of increasing complexity. With proteins we studied the compatibility of the tyrosine click reaction with cysteine and lysine-targeted labeling approaches and demonstrate that chemoselective trifunctionalization of proteins is readily achieved. In particular cases, we noted that PTAD decompn. resulted in formation of a putative isocyanate byproduct that was promiscuous in labeling. This side reaction product, however, was readily scavenged by the addn. of a small amt. of 2-amino-2-hydroxymethyl-propane-1,3-diol (Tris) to the reaction medium. To study the potential of the tyrosine click reaction to introduce poly(ethylene glycol) chains onto proteins (PEGylation), we demonstrate that this novel reagent provides for the selective PEGylation of chymotrypsinogen, whereas traditional succinimide-based PEGylation targeting lysine residues provided a more diverse range of PEGylated products. Finally, we applied the tyrosine click reaction to create a novel antibody-drug conjugate. For this purpose, we synthesized a PTAD deriv. linked to the HIV entry inhibitor aplaviroc. Labeling of the antibody trastuzumab with this reagent provided a labeled antibody conjugate that demonstrated potent HIV-1 neutralization activity demonstrating the potential of this reaction in creating protein conjugates with small mols. The tyrosine click linkage demonstrated stability to extremes of pH, temp., and exposure to human blood plasma indicating that this linkage is significantly more robust than maleimide-type linkages that are commonly employed in bioconjugations. These studies support the broad utility of this reaction in the chemoselective modification of small mols., peptides, and proteins under mild aq. conditions over a broad pH range using a wide variety of biol. acceptable buffers such as phosphate buffered saline (PBS) and 2-amino-2-hydroxymethyl-propane-1,3-diol (Tris) buffers as well as others and mixed buffered compns.
- 16Minamihata, K., Goto, M., and Kamiya, N. (2011) Site-specific protein cross-linking by peroxidase-catalyzed activation of a tyrosine-containing peptide tag Bioconjugate Chem. 22, 74– 81 DOI: 10.1021/bc1003982Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsFGhs7nM&md5=ddc68d94c1e48f91313aa366eefc50e9Site-Specific Protein Cross-Linking by Peroxidase-Catalyzed Activation of a Tyrosine-Containing Peptide TagMinamihata, Kosuke; Goto, Masahiro; Kamiya, NorihoBioconjugate Chemistry (2011), 22 (1), 74-81CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Protein modification methods represent fundamental techniques that are applicable in many fields. In this study, a site-specific protein crosslinking based on the oxidative tyrosine coupling reaction was demonstrated. In the presence of horseradish peroxidase (HRP) and H2O2, tyrosine residues undergo one-electron oxidn. reactions and form radicals in their phenolic moieties, and these species subsequently react with each other to form dimers or further react to generate polymers. Here, a peptide-tag contg. a tyrosine residue(s) (Y-tag, of which the amino acid sequences were either GGGGY or GGYYY) was genetically introduced at the C-terminus of a model protein, Escherichia coli alk. phosphatase (BAP). Following the incubation of recombinant BAPs with HRP and H2O2, Y-tagged BAPs were efficiently cross-linked with each other, whereas wild-type BAP did not undergo crosslinking, indicating that the tyrosine residues in the Y-tags were recognized by HRP as the substrates. To det. the site-specificity of the crosslinking reaction, the Y-tag was selectively removed by thrombin digestion. The resultant BAP without the Y-tag showed no reactivity in the presence of HRP and H2O2. Conversely, Y-tagged BAPs cross-linked by HRP treatment were almost completely digested into monomeric BAP units following incubation with the protease. Moreover, cross-linked Y-tagged BAPs retained ∼95% of their native enzymic activity. These results show that HRP catalyzed the site-specific crosslinking of BAPs through tyrosine residues positioned in the C-terminal Y-tag. The site-selective enzymic oxidative tyrosine coupling reaction should offer a practical option for site-specific and covalent protein modifications.
- 17Tilley, S. D. and Francis, M. B. (2006) Tyrosine-selective protein alkylation using pi-allylpalladium complexes J. Am. Chem. Soc. 128, 1080– 81 DOI: 10.1021/ja057106kGoogle Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XitVymsw%253D%253D&md5=8881b14279e96462847e868e6ede7c7eTyrosine-Selective Protein Alkylation Using π-Allylpalladium ComplexesTilley, S. David; Francis, Matthew B.Journal of the American Chemical Society (2006), 128 (4), 1080-1081CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)A new protein modification reaction has been developed based on a palladium-catalyzed allylic alkylation of tyrosine residues. This technique employs electrophilic π-allyl intermediates derived from allylic acetate and carbamate precursors and can be used to modify proteins in aq. soln. at room temp. To facilitate the detection of modified proteins using SDS-PAGE anal., a fluorescent allyl acetate was synthesized and coupled to chymotrypsinogen A and bacteriophage MS2. The tyrosine selectivity of the reaction was confirmed through trypsin digest anal. The utility of the reaction was demonstrated by using taurine-derived carbamates as water solubilizing groups that are cleaved upon protein functionalization. This soly. switching technique was used to install hydrophobic farnesyl and C17 chains on chymotrypsinogen A in water using little or no cosolvent. Following this, the C17 alkylated proteins were found to assoc. with lipid vesicles. In addn. to providing a new protein modification strategy targeting an underutilized amino acid side chain, this method provides convenient access to synthetic lipoproteins.
- 18Romanini, D. W. and Francis, M. B. (2008) Attachment of peptide building blocks to proteins through tyrosine bioconjugation Bioconjugate Chem. 19, 153– 7 DOI: 10.1021/bc700231vGoogle Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhsVWgtbbF&md5=e66a855c3a122ae3ea53db9e7d0c9be3Attachment of Peptide Building Blocks to Proteins Through Tyrosine BioconjugationRomanini, Dante W.; Francis, Matthew B.Bioconjugate Chemistry (2008), 19 (1), 153-157CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Recent efforts have yielded a no. of short peptide sequences with useful binding, sensing, and cellular uptake properties. To attach these sequences to tyrosine residues on intact proteins, a three-component Mannich-type strategy is reported. Two solid phase synthetic routes were developed to access peptides up to 20 residues in length with anilines at either the N- or C-termini. In the presence of 20 mM formaldehyde, these functional groups were coupled to tyrosine residues on proteins under mild reaction conditions. The identities of the resulting bioconjugates were confirmed using mass spectrometry and immunoblot anal. Screening expts. have demonstrated that the method is compatible with substrates contg. all of the amino acids, including lysine and cysteine residues. Importantly, tyrosine residues on proteins exhibit much faster reaction rates, allowing short peptides contg. this residue to be coupled without cross reactions.
- 19Long, M. J. C. and Hedstrom, L. (2012) Mushroom tyrosinase oxidizes tyrosine-rich sequences to allow selective protein functionalization ChemBioChem 13, 1818– 25 DOI: 10.1002/cbic.201100792Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtVCjtL7O&md5=0ad720d91299fcbef8304da0cee16763Mushroom Tyrosinase Oxidizes Tyrosine-Rich Sequences to Allow Selective Protein FunctionalizationLong, Marcus J. C.; Hedstrom, LizbethChemBioChem (2012), 13 (12), 1818-1825, S1818/1-S1818/12CODEN: CBCHFX; ISSN:1439-4227. (Wiley-VCH Verlag GmbH & Co. KGaA)We show that mushroom tyrosinase catalyzes the formation of reactive o-quinones on unstructured, tyrosine-rich sequences such as hemagglutinin (HA) tags (YPYDVPDYA). In the absence of exogenous nucleophiles and at low protein concns., the o-quinone decomps. with fragmentation of the HA tag. At higher protein concns. (>5 mg mL-1), crosslinking is obsd. Besthorn's reagent intercepts the o-quinone to give a characteristic pink complex that can be obsd. directly on a denaturing SDS-PAGE gel. Similar labeled species can be formed by using other nucleophiles such as Cy5-hydrazide. These reactions are selective for proteins bearing HA and other unstructured poly-tyrosine-contg. tags and can be performed in lysates to create specifically tagged proteins.
- 20Faccio, G., Kampf, M. M., Piatti, C., Thony-Meyer, L., and Richter, M. (2014) Tyrosinase-catalyzed site-specific immobilization of engineered C-phycocyanin to surface Sci. Rep. 4, 5370 DOI: 10.1038/srep05370Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXktlOjt7s%253D&md5=0e61c1970459d54a9709f6a30c0ea080Tyrosinase-catalyzed site-specific immobilization of engineered C-phycocyanin to surfaceFaccio, Greta; Kampf, Michael M.; Piatti, Chiara; Thony-Meyer, Linda; Richter, MichaelScientific Reports (2014), 4 (), 5370CODEN: SRCEC3; ISSN:2045-2322. (Nature Publishing Group)Enzymic crosslinking of proteins is often limited by the steric availability of the target residues, as of tyrosyl side chains in the case of tyrosinase. Carrying an N-terminal peptide-tag contg. two tyrosine residues, the fluorescent protein C-phycocyanin HisCPC from Synechocystis sp. PCC6803 was crosslinked to fluorescent high-mol. wt. forms with tyrosinase. Crosslinking with tyrosinase in the presence of L-tyrosine produced non fluorescent high-mol. wt. products. Incubated in the presence of tyrosinase, HisCPC could also be immobilized to amino-modified polystyrene beads thus conferring a blue fluorescence. Crosslinking and immobilization were site-specific as both processes required the presence of the N-terminal peptide in HisCPC.
- 21Ito, S., Kato, T., Shinpo, K., and Fujita, K. (1984) Oxidation of tyrosine residues in proteins by tyrosinase. Formation of protein-bonded 3,4-dihydroxyphenylalanine and 5-S-cysteinyl-3,4-dihydroxyphenylalanine Biochem. J. 222, 407– 11 DOI: 10.1042/bj2220407Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL2cXlsVOhtLo%253D&md5=c4d30cd7e9a3a9e217ceb2b17cf8d21dOxidation of tyrosine residues in proteins by tyrosinase. Formation of protein-bonded 3,4-dihydroxyphenylalanine and 5-S-cysteinyl-3,4-dihydroxyphenylalanineIto, Shosuke; Kato, Toshiaki; Shinpo, Kan; Fujita, KeisukeBiochemical Journal (1984), 222 (2), 407-11CODEN: BIJOAK; ISSN:0264-6021.A simple and rapid method was developed for the detn. of 3,4-dihydroxyphenylalanine (DOPA) and 5-S-cysteinyl-3,4-dihydroxyphenylalanine (5-S-cysteinyl-DOPA) in proteins with the use of high-pressure liq. chromatog. With this method, it was demonstrated that mushroom tyrosinase can catalyze hydroxylation of tyrosine residues in proteins to DOPA and subsequent oxidn. to dopaquinone residues. The dopaquinone residues in proteins combine with cysteine residues to form 5-S-cysteinyl-DOPA in bovine serum albumin and yeast alc. dehydrogenase, whereas DOPA is the major product in bovine insulin which lacks cysteine residues.
- 22Tabakovic, K. and Abul-Hajj, Y. J. (1994) Reaction of lysine with estrone 3,4-o-quinone Chem. Res. Toxicol. 7, 696– 701 DOI: 10.1021/tx00041a016Google ScholarThere is no corresponding record for this reference.
- 23Xu, R., Huang, X., Morgan, T. D., Prakash, O., Kramer, K. J., and Hawley, M. D. (1996) Characterization of products from the reactions of N-acetyldopamine quinone with N-acetylhistidine Arch. Biochem. Biophys. 329, 56– 64 DOI: 10.1006/abbi.1996.0191Google ScholarThere is no corresponding record for this reference.
- 24Borrmann, A., Fatunsin, O., Dommerholt, J., Jonker, A. M., Lowik, D. W., van Hest, J. C., and van Delft, F. L. (2015) Strain-promoted oxidation-controlled cyclooctyne-1,2-quinone cycloaddition (SPOCQ) for fast and activatable protein conjugation Bioconjugate Chem. 26, 257– 61 DOI: 10.1021/bc500534dGoogle Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitFCrs7vK&md5=7b072746ff08db93e05ad0dac3ae0966Strain-Promoted Oxidation-Controlled Cyclooctyne-1,2-Quinone Cycloaddition (SPOCQ) for Fast and Activatable Protein ConjugationBorrmann, Annika; Fatunsin, Olumide; Dommerholt, Jan; Jonker, Anika M.; Loewik, Dennis W. P. M.; van Hest, Jan C. M.; van Delft, Floris L.Bioconjugate Chemistry (2015), 26 (2), 257-261CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)A main challenge in the area of bioconjugation is to devise reactions that are both activatable and fast. Here, we introduce a temporally controlled reaction between cyclooctynes and 1,2-quinones, induced by facile oxidn. of 1,2-catechols. This so-called strain-promoted oxidn.-controlled cyclooctyne-1,2-quinone cycloaddn. (SPOCQ) shows a remarkably high reaction rate when performed with bicyclononyne (BCN), out-competing the well-known cycloaddn. of azides and BCN by 3 orders of magnitude, thereby allowing a new level of orthogonality in protein conjugation.
- 25Jonker, A. M., Borrmann, A., van Eck, E. R., van Delft, F. L., Lowik, D. W., and van Hest, J. C. (2015) A fast and activatable cross-linking strategy for hydrogel formation Adv. Mater. 27, 1235– 40 DOI: 10.1002/adma.201404448Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitFamsbbI&md5=c6d61b6bfee10fb1566884006b36fd1cA Fast and Activatable Cross-Linking Strategy for Hydrogel FormationJonker, Anika M.; Borrmann, Annika; van Eck, Ernst R. H.; van Delft, Floris L.; Loewik, Dennis W. P. M.; van Hest, Jan C. M.Advanced Materials (Weinheim, Germany) (2015), 27 (7), 1235-1240CODEN: ADVMEW; ISSN:0935-9648. (Wiley-VCH Verlag GmbH & Co. KGaA)The study reports a fast and activatible crosslinking method for hydrogel formation using a a strain-promoted oxidn. controlled cyclooctyne-1,2-quinone cycloaddn.
- 26Dommerholt, J., Schmidt, S., Temming, R., Hendriks, L. J., Rutjes, F. P., van Hest, J. C., Lefeber, D. J., Friedl, P., and van Delft, F. L. (2010) Readily accessible bicyclononynes for bioorthogonal labeling and three-dimensional imaging of living cells Angew. Chem., Int. Ed. 49, 9422– 5 DOI: 10.1002/anie.201003761Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsFSls73M&md5=1d18345acd06c8b7042111caccfdca06Readily accessible bicyclononynes for bioorthogonal labeling and three-dimensional imaging of living cellsDommerholt, Jan; Schmidt, Samuel; Temming, Rinske; Hendriks, Linda J. A.; Rutjes, Floris P. J. T.; van Hest, Jan C. M.; Lefeber, Dirk J.; Friedl, Peter; van Delft, Floris L.Angewandte Chemie, International Edition (2010), 49 (49), 9422-9425, S9422/1-S9422/22CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)We reported to use bicyclo[6.1.0]nonyne (BCN) as a novel ring-strained alkyne for metal-free cycloaddn. reactions with azides and nitrones. The complexes were synthesized and used for bioorthogonal labeling/three-dimensional imaging of living cells.
- 27Przybysz, A., Volmer, A. A., Westphal, A. H., and van Berkel, W. J. (2014) Bifunctional immobilization of a hyperthermostable endo-beta-1,3-glucanase Appl. Microbiol. Biotechnol. 98, 1155– 63 DOI: 10.1007/s00253-013-4953-3Google ScholarThere is no corresponding record for this reference.
- 28Wagner, K., Kwakkenbos, M. J., Claassen, Y. B., Maijoor, K., Bohne, M., van der Sluijs, K. F., Witte, M. D., van Zoelen, D. J., Cornelissen, L. A., and Beaumont, T. 2014, Bispecific antibody generated with sortase and click chemistry has broad antiinfluenza virus activity Proc. Natl. Acad. Sci. U. S. A. 111, 16820– 5 DOI: 10.1073/pnas.1408605111Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvV2gsLvN&md5=51a3e79489b81bc279bd38177c1c4d94Bispecific antibody generated with sortase and click chemistry has broad antiinfluenza virus activityWagner, Koen; Kwakkenbos, Mark J.; Claassen, Yvonne B.; Maijoor, Kelly; Boehne, Martino; van der Sluijs, Koenraad F.; Witte, Martin D.; van Zoelen, Diana J.; Cornelissen, Lisette A.; Beaumont, Tim; Bakker, Arjen Q.; Ploegh, Hidde L.; Spits, HergenProceedings of the National Academy of Sciences of the United States of America (2014), 111 (47), 16820-16825CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Bispecific antibodies have therapeutic potential by expanding the functions of conventional antibodies. Many different formats of bispecific antibodies have meanwhile been developed. Most are genetic modifications of the antibody backbone to facilitate incorporation of two different variable domains into a single mol. Here, we present a bispecific format where we have fused two full-sized IgG antibodies via their C termini using sortase trans-peptidation and click chem. to create a covalently linked IgG antibody heterodimer. By linking two potent anti-influenza A antibodies together, we have generated a full antibody dimer with bispecific activity that retains the activity and stability of the two fusion partners.
- 29Dorywalska, M., Strop, P., Melton-Witt, J. A., Hasa-Moreno, A., Farias, S. E., Galindo Casas, M., Delaria, K., Lui, V., Poulsen, K., and Loo, C. 2015, Effect of Attachment Site on Stability of Cleavable Antibody Drug Conjugates Bioconjugate Chem. 26, 650– 659 DOI: 10.1021/bc5005747Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvFGnsb4%253D&md5=c6ea72edbc33b0ad4aa743bc21707401Effect of Attachment Site on Stability of Cleavable Antibody Drug ConjugatesDorywalska, Magdalena; Strop, Pavel; Melton-Witt, Jody A.; Hasa-Moreno, Adela; Farias, Santiago E.; Galindo Casas, Meritxell; Delaria, Kathy; Lui, Victor; Poulsen, Kris; Loo, Carole; Krimm, Stellanie; Bolton, Gary; Moine, Ludivine; Dushin, Russell; Tran, Thomas-Toan; Liu, Shu-Hui; Rickert, Mathias; Foletti, Davide; Shelton, David L.; Pons, Jaume; Rajpal, ArvindBioconjugate Chemistry (2015), 26 (4), 650-659CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)The systemic stability of the antibody-drug linker is crucial for delivery of an intact antibody-drug conjugate (ADC) to target-expressing tumors. Linkers stable in circulation but readily processed in the target cell are necessary for both safety and potency of the delivered conjugate. Here, we report a range of stabilities for an auristatin-based payload site-specifically attached through a cleavable valine-citrulline-p-aminobenzylcarbamate (VC-PABC) linker across various sites on an antibody. We demonstrate that the conjugation site plays an important role in detg. VC-PABC linker stability in mouse plasma, and that the stability of the linker pos. correlates with ADC cytotoxic potency both in vitro and in vivo. Furthermore, we show that the VC-PABC cleavage in mouse plasma is not mediated by Cathepsin B, the protease thought to be primarily responsible for linker processing in the lysosomal degrdn. pathway. Although the VC-PABC cleavage is not detected in primate plasma in vitro, linker stabilization in the mouse is an essential prerequisite for designing successful efficacy and safety studies in rodents during preclin. stages of ADC programs. The divergence of linker metab. in mouse plasma and its intracellular cleavage offers an opportunity for linker optimization in the circulation without compromising its efficient payload release in the target cell.
- 30Chari, R. V., Miller, M. L., and Widdison, W. C. (2014) Antibody-drug conjugates: an emerging concept in cancer therapy Angew. Chem., Int. Ed. 53, 3796– 827 DOI: 10.1002/anie.201307628Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXjtVGmsr0%253D&md5=fbfb2f843f516654ea7c0eb98949604cAntibody-Drug Conjugates: An Emerging Concept in Cancer TherapyChari, Ravi V. J.; Miller, Michael L.; Widdison, Wayne C.Angewandte Chemie, International Edition (2014), 53 (15), 3796-3827CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Traditional cancer chemotherapy is often accompanied by systemic toxicity to the patient. Monoclonal antibodies against antigens on cancer cells offer an alternative tumor-selective treatment approach. However, most monoclonal antibodies are not sufficiently potent to be therapeutically active on their own. Antibody-drug conjugates (ADCs) use antibodies to deliver a potent cytotoxic compd. selectively to tumor cells, thus improving the therapeutic index of chemotherapeutic agents. The recent approval of two ADCs, brentuximab vedotin and ado-trastuzumab emtansine, for cancer treatment has spurred tremendous research interest in this field. This Review touches upon the early efforts in the field, and describes how the lessons learned from the first-generation ADCs have led to improvements in every aspect of this technol., i.e., the antibody, the cytotoxic compd., and the linker connecting them, leading to the current successes. The design of ADCs currently in clin. development, and results from mechanistic studies and preclin. and clin. evaluation are discussed. Emerging technologies that seek to further advance this exciting area of research are also discussed.
- 31Younes, A., Bartlett, N. L., Leonard, J. P., Kennedy, D. A., Lynch, C. M., Sievers, E. L., and Forero-Torres, A. (2010) Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas N. Engl. J. Med. 363, 1812– 21 DOI: 10.1056/NEJMoa1002965Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtl2gsbbE&md5=d27d22f971143a6c5415090ab57fe01aBrentuximab vedotin (SGN-35) for relapsed Cd30-positive lymphomasYounes, Anas; Bartlett, Nancy L.; Leonard, John P.; Kennedy, Dana A.; Lynch, Carmel M.; Sievers, Eric L.; Forero-Torres, AndresNew England Journal of Medicine (2010), 363 (19), 1812-1821CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)BACKGROUND: Hodgkin's lymphoma and anaplastic large-cell lymphoma are the two most common tumors expressing CD30. Previous attempts to target the CD30 antigen with monoclonal-based therapies have shown minimal activity. To enhance the antitumor activity of CD30-directed therapy, the antitubulin agent monomethyl auristatin E (MMAE) was attached to a CD30-specific monoclonal antibody by an enzyme-cleavable linker, producing the antibody-drug conjugate brentuximab vedotin (SGN-35). METHODS: In this phase 1, open-label, multicenter dose-escalation study, we administered brentuximab vedotin (at a dose of 0.1 to 3.6 mg per kg of body wt.) every 3 wk to 45 patients with relapsed or refractory CD30-pos. hematol. Cancers, primarily Hodgkin's lymphoma and anaplastic large-cell lymphoma. Patients had received a median of three previous chemotherapy regimens (range, one to seven), and 73% had undergone autologous stem-cell transplantation. RESULTS: The max. tolerated dose was 1.8 mg per kg, administered every 3 wk. Objective responses, including 11 complete remissions, were obsd. in 17 patients. Of 12 patients who received the 1.8-mg-per-kilogram dose, 6 (50%) had an objective response. The median duration of response was at least 9.7 mo. Tumor regression was obsd. in 36 of 42 patients who could be evaluated (86%). The most common adverse events were fatigue, pyrexia, diarrhea, nausea, neutropenia, and peripheral neuropathy. CONCLUSIONS: Brentuximab vedotin induced durable objective responses and resulted in tumor regression for most patients with relapsed or refractory CD30-pos. lymphomas in this phase 1 study. Treatment was assocd. primarily with grade 1 or 2 (mild-to-moderate) toxic effects.
- 32Verma, S., Miles, D., Gianni, L., Krop, I. E., Welslau, M., Baselga, J., Pegram, M., Oh, D. Y., Dieras, V., and Guardino, E. 2012, Trastuzumab emtansine for HER2-positive advanced breast cancer N. Engl. J. Med. 367, 1783– 91 DOI: 10.1056/NEJMoa1209124Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhs1ekt73M&md5=3be5bac27a81b530442dc8a6b4a1f82eTrastuzumab emtansine for HER2-positive advanced breast cancerVerma, Sunil; Miles, David; Gianni, Luca; Krop, Ian E.; Welslau, Manfred; Baselga, Jose; Pegram, Mark; Oh, Do-Youn; Dieras, Veronique; Guardino, Ellie; Fang, Liang; Lu, Michael W.; Olsen, Steven; Blackwell, KimNew England Journal of Medicine (2012), 367 (19), 1783-1791CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)BACKGROUND: Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate incorporating the human epidermal growth factor receptor 2 (HER2)-targeted antitumor properties of trastuzumab with the cytotoxic activity of the microtubule-inhibitory agent DM1. The antibody and the cytotoxic agent are conjugated by means of a stable linker. METHODS: We randomly assigned patients with HER2-pos. advanced breast cancer, who had previously been treated with trastuzumab and a taxane, to T-DM1 or lapatinib plus capecitabine. The primary end points were progression-free survival (as assessed by independent review), overall survival, and safety. Secondary end points included progression-free survival (investigator-assessed), the objective response rate, and the time to symptom progression. Two interim analyses of overall survival were conducted. RESULTS: Among 991 randomly assigned patients, median progression-free survival as assessed by independent review was 9.6 mo with T-DM1 vs. 6.4 mo with lapatinib plus capecitabine (hazard ratio for progression or death from any cause, 0.65; 95% confidence interval [CI], 0.55 to 0.77; P<0.001), and median overall survival at the second interim anal. crossed the stopping boundary for efficacy (30.9 mo vs. 25.1 mo; hazard ratio for death from any cause, 0.68; 95% CI, 0.55 to 0.85; P<0.001). The objective response rate was higher with T-DM1 (43.6%, vs. 30.8% with lapatinib plus capecitabine; P<0.001); results for all addnl. secondary end points favored T-DM1. Rates of grade 3 or above were higher with lapatinib plus capecitabine than with T-DM1 (57% vs. 41%). The incidences of thrombocytopenia and increased serum aminotransferase levels were higher with T-DM1, whereas the incidences of diarrhea, nausea, vomiting, and palmar-plantar erythrodysesthesia were higher with lapatinib plus capecitabine. CONCLUSIONS: T-DM1 significantly prolonged progression-free and overall survival with less toxicity than lapatinib plus capecitabine in patients with HER2-pos. advanced breast cancer previously treated with trastuzumab and a taxane.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 77 publications.
- Irene Shajan, Léa N. C. Rochet, Shannon R. Tracey, Rania Benazza, Bianka Jackowska, Oscar Hernandez-Alba, Sarah Cianférani, Christopher J. Scott, Floris L. van Delft, Vijay Chudasama, Bauke Albada. Modular Semisynthetic Approach to Generate T Cell-Dependent Bispecific Constructs from Recombinant IgG1 Antibodies. Bioconjugate Chemistry 2024, 35
(10)
, 1524-1531. https://doi.org/10.1021/acs.bioconjchem.4c00309
- Saki Horie, Kenji Mishiro, Mio Nishino, Inori Domae, Mitsuo Wakasugi, Tsukasa Matsunaga, Munetaka Kunishima. Epitope-Based Specific Antibody Modifications. Bioconjugate Chemistry 2023, 34
(11)
, 2022-2033. https://doi.org/10.1021/acs.bioconjchem.3c00340
- Aaron Debon, Elina Siirola, Radka Snajdrova. Enzymatic Bioconjugation: A Perspective from the Pharmaceutical Industry. JACS Au 2023, 3
(5)
, 1267-1283. https://doi.org/10.1021/jacsau.2c00617
- 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
- Julia E. Rosenberger, Yixin Xie, Yinzhi Fang, Xinyi Lyu, William S. Trout, Olga Dmitrenko, Joseph M. Fox. Ligand-Directed Photocatalysts and Far-Red Light Enable Catalytic Bioorthogonal Uncaging inside Live Cells. Journal of the American Chemical Society 2023, 145
(11)
, 6067-6078. https://doi.org/10.1021/jacs.2c10655
- Johnathan C. Maza, Derek M. García-Almedina, Lydia E. Boike, Noah X. Hamlish, Daniel K. Nomura, Matthew B. Francis. Tyrosinase-Mediated Synthesis of Nanobody–Cell Conjugates. ACS Central Science 2022, 8
(7)
, 955-962. https://doi.org/10.1021/acscentsci.1c01265
- Andrew Jemas, Yixin Xie, Jessica E. Pigga, Jeffrey L. Caplan, Christopher W. am Ende, Joseph M. Fox. Catalytic Activation of Bioorthogonal Chemistry with Light (CABL) Enables Rapid, Spatiotemporally Controlled Labeling and No-Wash, Subcellular 3D-Patterning in Live Cells Using Long Wavelength Light. Journal of the American Chemical Society 2022, 144
(4)
, 1647-1662. https://doi.org/10.1021/jacs.1c10390
- Katsuya Maruyama, Takashi Ishiyama, Yohei Seki, Kentaro Sakai, Takaya Togo, Kounosuke Oisaki, Motomu Kanai. Protein Modification at Tyrosine with Iminoxyl Radicals. Journal of the American Chemical Society 2021, 143
(47)
, 19844-19855. https://doi.org/10.1021/jacs.1c09066
- Jorick J. Bruins, Johannes A. M. Damen, Marloes A. Wijdeven, Lianne P. W. M. Lelieveldt, Floris L. van Delft, Bauke Albada. Non-Genetic Generation of Antibody Conjugates Based on Chemoenzymatic Tyrosine Click Chemistry. Bioconjugate Chemistry 2021, 32
(10)
, 2167-2172. https://doi.org/10.1021/acs.bioconjchem.1c00351
- Casey S. Mogilevsky, Marco J. Lobba, Daniel D. Brauer, Alan M. Marmelstein, Johnathan C. Maza, Jamie M. Gleason, Jennifer A. Doudna, Matthew B. Francis. Synthesis of Multi-Protein Complexes through Charge-Directed Sequential Activation of Tyrosine Residues. Journal of the American Chemical Society 2021, 143
(34)
, 13538-13547. https://doi.org/10.1021/jacs.1c03079
- 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
- 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
- Bauke Albada, Jordi F. Keijzer, Han Zuilhof, Floris van Delft. Oxidation-Induced “One-Pot” Click Chemistry. Chemical Reviews 2021, 121
(12)
, 7032-7058. https://doi.org/10.1021/acs.chemrev.0c01180
- Napon Nilchan, James M. Alburger, William R. Roush, Christoph Rader. An Engineered Arginine Residue of Unusual pH-Sensitive Reactivity Facilitates Site-Selective Antibody Conjugation. Biochemistry 2021, 60
(14)
, 1080-1087. https://doi.org/10.1021/acs.biochem.0c00955
- Jordi F. Keijzer, Bauke Albada. Site-Specific and Trigger-Activated Modification of Proteins by Means of Catalytic Hemin/G-quadruplex DNAzyme Nanostructures. Bioconjugate Chemistry 2020, 31
(10)
, 2283-2287. https://doi.org/10.1021/acs.bioconjchem.0c00422
- Marco J. Lobba, Christof Fellmann, Alan M. Marmelstein, Johnathan C. Maza, Elijah N. Kissman, Stephanie A. Robinson, Brett T. Staahl, Cole Urnes, Rachel J. Lew, Casey S. Mogilevsky, Jennifer A. Doudna, Matthew B. Francis. Site-Specific Bioconjugation through Enzyme-Catalyzed Tyrosine–Cysteine Bond Formation. ACS Central Science 2020, 6
(9)
, 1564-1571. https://doi.org/10.1021/acscentsci.0c00940
- Shinichi Sato, Masaki Matsumura, Tetsuya Kadonosono, Satoshi Abe, Takafumi Ueno, Hiroshi Ueda, Hiroyuki Nakamura. Site-Selective Protein Chemical Modification of Exposed Tyrosine Residues Using Tyrosine Click Reaction. Bioconjugate Chemistry 2020, 31
(5)
, 1417-1424. https://doi.org/10.1021/acs.bioconjchem.0c00120
- Alan M. Marmelstein, Marco J. Lobba, Casey S. Mogilevsky, Johnathan C. Maza, Daniel D. Brauer, Matthew B. Francis. Tyrosinase-Mediated Oxidative Coupling of Tyrosine Tags on Peptides and Proteins. Journal of the American Chemical Society 2020, 142
(11)
, 5078-5086. https://doi.org/10.1021/jacs.9b12002
- Jorick
J. Bruins, Criss van de Wouw, Koen Wagner, Lina Bartels, Bauke Albada, Floris L. van Delft. Highly Efficient Mono-Functionalization of Knob-in-Hole Antibodies with Strain-Promoted Click Chemistry. ACS Omega 2019, 4
(7)
, 11801-11807. https://doi.org/10.1021/acsomega.9b01727
- Johnathan
C. Maza, Daniel L. V. Bader, Lifeng Xiao, Alan M. Marmelstein, Daniel D. Brauer, Adel M. ElSohly, Matthew J. Smith, Shane W. Krska, Craig A. Parish, Matthew B. Francis. Enzymatic Modification of N-Terminal Proline Residues Using Phenol Derivatives. Journal of the American Chemical Society 2019, 141
(9)
, 3885-3892. https://doi.org/10.1021/jacs.8b10845
- Elita Montanari, Arianna Gennari, Maria Pelliccia, Lucio Manzi, Roberto Donno, Neil J. Oldham, Andrew MacDonald, Nicola Tirelli. Tyrosinase-Mediated Bioconjugation. A Versatile Approach to Chimeric Macromolecules. Bioconjugate Chemistry 2018, 29
(8)
, 2550-2560. https://doi.org/10.1021/acs.bioconjchem.8b00227
- Yi Liu, Hsuan-Chen Wu, Narendranath Bhokisham, Jinyang Li, Kai-Lin Hong, David N. Quan, Chen-Yu Tsao, William E. Bentley, Gregory F. Payne. Biofabricating Functional Soft Matter Using Protein Engineering to Enable Enzymatic Assembly. Bioconjugate Chemistry 2018, 29
(6)
, 1809-1822. https://doi.org/10.1021/acs.bioconjchem.8b00197
- Jorge Escorihuela, Anita Das, Wilhelmus J. E. Looijen, Floris L. van Delft, Adelia J. A. Aquino, Hans Lischka, and Han Zuilhof . Kinetics of the Strain-Promoted Oxidation-Controlled Cycloalkyne-1,2-quinone Cycloaddition: Experimental and Theoretical Studies. The Journal of Organic Chemistry 2018, 83
(1)
, 244-252. https://doi.org/10.1021/acs.joc.7b02614
- Matthias Pretzler, Annette Rompel. Tyrosinases: a family of copper-containing metalloenzymes. ChemTexts 2024, 10
(4)
https://doi.org/10.1007/s40828-024-00195-y
- Ming Bao, Klaudia Łuczak, Wojciech Chaładaj, Marriah Baird, Dorota Gryko, Michael P. Doyle. Photo-cycloaddition reactions of vinyldiazo compounds. Nature Communications 2024, 15
(1)
https://doi.org/10.1038/s41467-024-48274-5
- Keita Nakane, Chizu Fujimura, Shogo Miyano, Zhengyi Liu, Tatsuya Niwa, Hafumi Nishi, Tetsuya Kadonosono, Hideki Taguchi, Shusuke Tomoshige, Minoru Ishikawa, Shinichi Sato. Laccase-catalyzed tyrosine click reaction with 1-methyl-4-arylurazole: rapid labeling on protein surfaces. Chemical Communications 2024, 60
(96)
, 14208-14211. https://doi.org/10.1039/D4CC03802A
- Kevin R. Venrooij, Lucienne de Bondt, Kimberly M. Bonger. Mutually Orthogonal Bioorthogonal Reactions: Selective Chemistries for Labeling Multiple Biomolecules Simultaneously. Topics in Current Chemistry 2024, 382
(3)
https://doi.org/10.1007/s41061-024-00467-8
- Hongfei Chen, Hong‐Chai Fabio Wong, Jiaming Qiu, Biquan Li, Dingdong Yuan, Hao Kong, Yishu Bao, Yu Zhang, Zhiyi Xu, Ying‐Lung Steve Tse, Jiang Xia. Site‐Selective Tyrosine Reaction for Antibody‐Cell Conjugation and Targeted Immunotherapy. Advanced Science 2024, 11
(5)
https://doi.org/10.1002/advs.202305012
- Thomas A. King, Laura Rodríguez Pérez, Sabine L. Flitsch. Application of Biocatalysis for Protein Bioconjugation. 2024, 389-437. https://doi.org/10.1016/B978-0-32-390644-9.00122-0
- Christopher C. Marvin, Milan Bruncko, Ippei Usui. Chemistry of Antibody-Small Molecule Drug Conjugates. 2024https://doi.org/10.1016/B978-0-323-96025-0.00109-5
- Hwaseok Hong, Uk-Jae Lee, Seul Hoo Lee, Hyun Kim, Gyu-Min Lim, Sang-Hyuk Lee, Hyeoncheol Francis Son, Byung-Gee Kim, Kyung-Jin Kim. Highly efficient site-specific protein modification using tyrosinase from Streptomyces avermitilis: Structural insight. International Journal of Biological Macromolecules 2024, 255 , 128313. https://doi.org/10.1016/j.ijbiomac.2023.128313
- Xiaoxuan Ma, Jian Jiang, Xiaoye An, Wanting Zu, Chi Ma, Zhuo Zhang, Yaci Lu, Lijing Zhao, Lisheng Wang. Advances in research based on antibody-cell conjugation. Frontiers in Immunology 2023, 14 https://doi.org/10.3389/fimmu.2023.1310130
- Valentina Vitali, Francesco Torricella, Lara Massai, Luigi Messori, Lucia Banci. Enlarging the scenario of site directed 19F labeling for NMR spectroscopy of biomolecules. Scientific Reports 2023, 13
(1)
https://doi.org/10.1038/s41598-023-49247-2
- Bauke Albada. Precise and Controlled Modification of Proteins using Multifunctional Chemical Constructs. ChemBioChem 2023, 24
(15)
https://doi.org/10.1002/cbic.202300187
- Shengping Zhang, Luis M. De Leon Rodriguez, Freda F. Li, Margaret A. Brimble. Recent developments in the cleavage, functionalization, and conjugation of proteins and peptides at tyrosine residues. Chemical Science 2023, 14
(29)
, 7782-7817. https://doi.org/10.1039/D3SC02543H
- Johannes A. M. Damen, Jorge Escorihuela, Han Zuilhof, Floris L. van Delft, Bauke Albada. High Rates of Quinone‐Alkyne Cycloaddition Reactions are Dictated by Entropic Factors. Chemistry – A European Journal 2023, 29
(39)
https://doi.org/10.1002/chem.202300231
- Ryan J. Pakula, Peter J. H. Scott. Applications of radiolabeled antibodies in neuroscience and neuro‐oncology. Journal of Labelled Compounds and Radiopharmaceuticals 2023, 66
(9)
, 269-285. https://doi.org/10.1002/jlcr.4049
- Niklas Henrik Fischer, Maria Teresa Oliveira, Frederik Diness. Chemical modification of proteins – challenges and trends at the start of the 2020s. Biomaterials Science 2023, 11
(3)
, 719-748. https://doi.org/10.1039/D2BM01237E
- Shunsuke Yamazaki, Yutaka Matsuda. Tag‐Free Enzymatic Modification for Antibody−Drug Conjugate Production. ChemistrySelect 2022, 7
(48)
https://doi.org/10.1002/slct.202203753
- Hao‐Ran Jia, Ya‐Xuan Zhu, Yi Liu, Yuxin Guo, Sayed Mir Sayed, Xiao‐Yu Zhu, Xiaotong Cheng, Fu‐Gen Wu. Direct chemical editing of Gram‐positive bacterial cell walls via an enzyme‐catalyzed oxidative coupling reaction. Exploration 2022, 2
(5)
https://doi.org/10.1002/EXP.20220010
- Jordi F. Keijzer, Han Zuilhof, Bauke Albada. Calibrating Catalytic DNA Nanostructures for Site‐Selective Protein Modification**. Chemistry – A European Journal 2022, 28
(51)
https://doi.org/10.1002/chem.202200895
- Huaibin Yu, Jiayi Feng, Fangrui Zhong, Yuzhou Wu. Chemical Modification for the “Off‐/On” Regulation of Enzyme Activity. Macromolecular Rapid Communications 2022, 43
(18)
https://doi.org/10.1002/marc.202200195
- Pablo García-Aznar, Jorge Escorihuela. Computational insights into the inverse electron-demand Diels–Alder reaction of norbornenes with 1,2,4,5-tetrazines: norbornene substituents’ effects on the reaction rate. Organic & Biomolecular Chemistry 2022, 20
(32)
, 6400-6412. https://doi.org/10.1039/D2OB01121B
- Hongfei Jiang, Qing Zhang, Yue Zhang, Huxin Feng, Hao Jiang, Fan Pu, Rilei Yu, Zheng Zhong, Chaoming Wang, Yi Man Eva Fung, Pilar Blasco, Yongxin Li, Tao Jiang, Xuechen Li. Triazine-pyridine chemistry for protein labelling on tyrosine. Chemical Communications 2022, 58
(50)
, 7066-7069. https://doi.org/10.1039/D2CC01528E
- Shengping Zhang, Luis M. De Leon Rodriguez, Freda F. Li, Renjie Huang, Ivanhoe K. H. Leung, Paul W. R. Harris, Margaret A. Brimble. A novel tyrosine hyperoxidation enables selective peptide cleavage. Chemical Science 2022, 13
(9)
, 2753-2763. https://doi.org/10.1039/D1SC06216F
- Jonathan Schwach, Mustafa Abdellatif, Andreas Stengl. More than Toxins—Current Prospects in Designing the Next Generation of Antibody Drug Conjugates. Frontiers in Bioscience-Landmark 2022, 27
(8)
https://doi.org/10.31083/j.fbl2708240
- G. T. Hermanson, F. L. van Delft. Antibody Conjugation Technologies. 2021, 32-70. https://doi.org/10.1039/9781839165153-00032
- Sébastien Depienne, Dimitri Alvarez-Dorta, Mikael Croyal, Ranil C. T. Temgoua, Cathy Charlier, David Deniaud, Mathieu Mével, Mohammed Boujtita, Sébastien G. Gouin. Luminol anchors improve the electrochemical-tyrosine-click labelling of proteins. Chemical Science 2021, 12
(46)
, 15374-15381. https://doi.org/10.1039/D1SC04809K
- Augustine George, Mohan Indhu, Sundarapandian Ashokraj, Ganesh Shanmugam, Ponesakki Ganesan, Numbi Ramudu Kamini, Niraikulam Ayyadurai. Genetically encoded dihydroxyphenylalanine coupled with tyrosinase for strain promoted labeling. Bioorganic & Medicinal Chemistry 2021, 50 , 116460. https://doi.org/10.1016/j.bmc.2021.116460
- Zhefu Dai, Xiao-Nan Zhang, Qinqin Cheng, Fan Fei, Tianling Hou, Jiawei Li, Alireza Abdolvahabi, Junji Watanabe, Hua Pei, Goar Smbatyan, Jianming Xie, Heinz-Josef Lenz, Stan G. Louie, Yong Zhang. Site-specific antibody-drug conjugates with variable drug-to-antibody-ratios for AML therapy. Journal of Controlled Release 2021, 336 , 433-442. https://doi.org/10.1016/j.jconrel.2021.06.041
- Vesela Kostova, Patrice Désos, Jérôme-Benoît Starck, Andras Kotschy. The Chemistry Behind ADCs. Pharmaceuticals 2021, 14
(5)
, 442. https://doi.org/10.3390/ph14050442
- Mike L.W.J. Smeenk, Jordi Agramunt, Kimberly M. Bonger. Recent developments in bioorthogonal chemistry and the orthogonality within. Current Opinion in Chemical Biology 2021, 60 , 79-88. https://doi.org/10.1016/j.cbpa.2020.09.002
- Stephen J. Walsh, Jonathan D. Bargh, Friederike M. Dannheim, Abigail R. Hanby, Hikaru Seki, Andrew J. Counsell, Xiaoxu Ou, Elaine Fowler, Nicola Ashman, Yuri Takada, Albert Isidro-Llobet, Jeremy S. Parker, Jason S. Carroll, David R. Spring. Site-selective modification strategies in antibody–drug conjugates. Chemical Society Reviews 2021, 50
(2)
, 1305-1353. https://doi.org/10.1039/D0CS00310G
- Peter A. Szijj, Kristina A. Kostadinova, Richard J. Spears, Vijay Chudasama. Tyrosine bioconjugation – an emergent alternative. Organic & Biomolecular Chemistry 2020, 18
(44)
, 9018-9028. https://doi.org/10.1039/D0OB01912G
- Dimitri Alvarez Dorta, David Deniaud, Mathieu Mével, Sébastien G. Gouin. Tyrosine Conjugation Methods for Protein Labelling. Chemistry – A European Journal 2020, 26
(63)
, 14257-14269. https://doi.org/10.1002/chem.202001992
- Sean S. Nguyen, Jennifer A. Prescher. Developing bioorthogonal probes to span a spectrum of reactivities. Nature Reviews Chemistry 2020, 4
(9)
, 476-489. https://doi.org/10.1038/s41570-020-0205-0
- Sara Ponziani, Giulia Di Vittorio, Giuseppina Pitari, Anna Maria Cimini, Matteo Ardini, Roberta Gentile, Stefano Iacobelli, Gianluca Sala, Emily Capone, David J. Flavell, Rodolfo Ippoliti, Francesco Giansanti. Antibody-Drug Conjugates: The New Frontier of Chemotherapy. International Journal of Molecular Sciences 2020, 21
(15)
, 5510. https://doi.org/10.3390/ijms21155510
- Zhefu Dai, Xiao-Nan Zhang, Fariborz Nasertorabi, Qinqin Cheng, Jiawei Li, Benjamin B. Katz, Goar Smbatyan, Hua Pei, Stan G. Louie, Heinz-Josef Lenz, Raymond C. Stevens, Yong Zhang. Synthesis of site-specific antibody-drug conjugates by ADP-ribosyl cyclases. Science Advances 2020, 6
(23)
https://doi.org/10.1126/sciadv.aba6752
- Hendrik Schneider, Lukas Deweid, Olga Avrutina, Harald Kolmar. Recent progress in transglutaminase-mediated assembly of antibody-drug conjugates. Analytical Biochemistry 2020, 595 , 113615. https://doi.org/10.1016/j.ab.2020.113615
- L. Nathan Tumey. An Overview of the Current ADC Discovery Landscape. 2020, 1-22. https://doi.org/10.1007/978-1-4939-9929-3_1
- Lukas Deweid, Olga Avrutina, Harald Kolmar. Microbial transglutaminase for biotechnological and biomedical engineering. Biological Chemistry 2019, 400
(3)
, 257-274. https://doi.org/10.1515/hsz-2018-0335
- Lina Bartels, Hidde L. Ploegh, Hergen Spits, Koen Wagner. Preparation of bispecific antibody-protein adducts by site-specific chemo-enzymatic conjugation. Methods 2019, 154 , 93-101. https://doi.org/10.1016/j.ymeth.2018.07.013
- Jorick J. Bruins, Criss van de Wouw, Jordi F. Keijzer, Bauke Albada, Floris L. van Delft. Inducible, Selective Labeling of Proteins via Enzymatic Oxidation of Tyrosine. 2019, 357-368. https://doi.org/10.1007/978-1-4939-9546-2_18
- Shino Manabe. Recent Progress in Linker Technology for Antibody-Drug Conjugates: Methods for Connection and Release. 2019, 93-123. https://doi.org/10.1007/978-4-431-56880-3_5
- Hiroyuki Nakamura. Target Protein Chemical Modification. 2019, 305-333. https://doi.org/10.1007/978-981-13-6244-6_13
- Sander S. van Berkel, Floris L. van Delft. Enzymatic strategies for (near) clinical development of antibody-drug conjugates. Drug Discovery Today: Technologies 2018, 30 , 3-10. https://doi.org/10.1016/j.ddtec.2018.09.005
- Si Mou, Yue Huang, Anton I. Rosenbaum. ADME Considerations and Bioanalytical Strategies for Pharmacokinetic Assessments of Antibody-Drug Conjugates. Antibodies 2018, 7
(4)
, 41. https://doi.org/10.3390/antib7040041
- Digvijay Gahtory, Rickdeb Sen, Andriy R. Kuzmyn, Jorge Escorihuela, Han Zuilhof. Strain‐Promoted Cycloaddition of Cyclopropenes with
o
‐Quinones: A Rapid Click Reaction. Angewandte Chemie 2018, 130
(32)
, 10275-10279. https://doi.org/10.1002/ange.201800937
- Digvijay Gahtory, Rickdeb Sen, Andriy R. Kuzmyn, Jorge Escorihuela, Han Zuilhof. Strain‐Promoted Cycloaddition of Cyclopropenes with
o
‐Quinones: A Rapid Click Reaction. Angewandte Chemie International Edition 2018, 57
(32)
, 10118-10122. https://doi.org/10.1002/anie.201800937
- Digvijay Gahtory, Rickdeb Sen, Sidharam Pujari, Suhua Li, Qinheng Zheng, John E. Moses, K. Barry Sharpless, Han Zuilhof. Quantitative and Orthogonal Formation and Reactivity of SuFEx Platforms. Chemistry – A European Journal 2018, 24
(41)
, 10550-10556. https://doi.org/10.1002/chem.201802356
- Jorick J. Bruins, Bauke Albada, Floris van Delft. ortho
‐Quinones and Analogues Thereof: Highly Reactive Intermediates for Fast and Selective Biofunctionalization. Chemistry – A European Journal 2018, 24
(19)
, 4749-4756. https://doi.org/10.1002/chem.201703919
- Jun Ohata, Mary K. Miller, Courtney M. Mountain, Farrukh Vohidov, Zachary T. Ball. A Three‐Component Organometallic Tyrosine Bioconjugation. Angewandte Chemie 2018, 130
(11)
, 2877-2880. https://doi.org/10.1002/ange.201711868
- Jun Ohata, Mary K. Miller, Courtney M. Mountain, Farrukh Vohidov, Zachary T. Ball. A Three‐Component Organometallic Tyrosine Bioconjugation. Angewandte Chemie International Edition 2018, 57
(11)
, 2827-2830. https://doi.org/10.1002/anie.201711868
- Olivier Marcq. Outlook on Next Generation Technologies and Strategy Considerations for ADC Process Development and Manufacturing. 2018, 113-161. https://doi.org/10.1007/978-3-319-78154-9_6
- Igor Dovgan, Stéphane Erb, Steve Hessmann, Sylvain Ursuegui, Chloé Michel, Christian Muller, Guilhem Chaubet, Sarah Cianférani, Alain Wagner. Arginine-selective bioconjugation with 4-azidophenyl glyoxal: application to the single and dual functionalisation of native antibodies. Organic & Biomolecular Chemistry 2018, 16
(8)
, 1305-1311. https://doi.org/10.1039/C7OB02844J
- Jorick J. Bruins, Daniel Blanco-Ania, Vincent van der Doef, Floris L. van Delft, Bauke Albada. Orthogonal, dual protein labelling by tandem cycloaddition of strained alkenes and alkynes to
ortho
-quinones and azides. Chemical Communications 2018, 54
(53)
, 7338-7341. https://doi.org/10.1039/C8CC02638F
- Alex R. Nanna, Xiuling Li, Even Walseng, Lee Pedzisa, Rebecca S. Goydel, David Hymel, Terrence R. Burke, William R. Roush, Christoph Rader. Harnessing a catalytic lysine residue for the one-step preparation of homogeneous antibody-drug conjugates. Nature Communications 2017, 8
(1)
https://doi.org/10.1038/s41467-017-01257-1
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
Abstract
Figure 1
Figure 1. SPOCQ labeling of G4Y-tagged laminarinase A by reaction of BCN-modified reagent 1 with in situ generated 1,2-quinone. Typical reaction conditions: LamA (1.0 mg/mL), mTyr (0.3 mg/mL), and 1 (4 equiv) in 50 mM potassium phosphate buffer pH 7.3, containing 135 mM NaCl and 10% DMSO as cosolvent.
Figure 2
Figure 2. (A) SDS-PAGE analysis of SPOCQ on LamA–G4Y and wt-LamA. (B) MS profile of LamA–G4Y. (C) MS profile of LamA–G4Y after SPOCQ with 1.
Figure 3
Figure 3. (A) Schematic representation of G4Y-tagged antibodies. (B) SDS-PAGE analysis of SPOCQ on AT1002[LC]G4Y and wt-AT1002. (C) MS profile of AT1002[LC]G4Y (light chain only). (D) MS spectrum of AT1002[LC]G4Y after SPOCQ with 1. (E) MS profile of AT1002[LC]G4Y after SPOCQ with 2.
References
This article references 32 other publications.
- 1Foley, T. L. and Burkart, M. D. (2007) Site-specific protein modification: advances and applications Curr. Opin. Chem. Biol. 11, 12– 19 DOI: 10.1016/j.cbpa.2006.11.0361https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhsFOhs70%253D&md5=b527edf8f4a8be8dbe10b4f36aa87937Site-specific protein modification: Advances and applicationsFoley, Timothy L.; Burkart, Michael D.Current Opinion in Chemical Biology (2007), 11 (1), 12-19CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)A review. Although chem. methods to modify proteins in a sequence-specific manner have yet to be developed, site-specific post-translational modification of proteins has recently emerged as a major focus in biol. chem. Post-translational modification with functionalized substrate analogs opens up several unique avenues to induce selective reactivity into proteins in a sequence-specific manner, and can be applied to protein identification and manipulation in both in vitro and in vivo contexts. Further in vivo applications of this method will enable the imaging of cellular processes, avoiding nonspecific labeling and probe scattering, major complications obsd. in nonenzymic methods. Addnl., new tools for in vitro protein modification have been developed that offer more versatile ways to study protein structure and function.
- 2Jung, S. and Kwon, I. (2016) Expansion of bioorthogonal chemistries towards site-specific polymer-protein conjugation Polym. Chem. 7, 4584– 98 DOI: 10.1039/C6PY00856A2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XpvFeisrw%253D&md5=d8ee4ce063caa8dd73b0ab48da2780b4Expansion of bioorthogonal chemistries towards site-specific polymer-protein conjugationJung, Secheon; Kwon, InchanPolymer Chemistry (2016), 7 (28), 4584-4598CODEN: PCOHC2; ISSN:1759-9962. (Royal Society of Chemistry)Polymer conjugation to proteins has been widely used to improve or expand protein properties. However, polymer conjugation to random sites of a target protein often led to a significant loss of crit. protein properties. In order to overcome this, polymer conjugation to specific sites of a protein was developed using the site-specific introduction of non-natural amino acids and bioorthogonal chemistries. This review summarizes the recent advances in bioorthogonal chemistries. As the repertoire of bioorthogonal chemistries available for polymer conjugation is expanding, the site-specific polymer conjugation technique would be a valuable platform technique to design novel protein-polymer conjugates.
- 3Spicer, C. D. and Davis, B. G. (2014) Selective chemical protein modification Nat. Commun. 5, 4740 DOI: 10.1038/ncomms57403https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXksVeksb4%253D&md5=055c882ff0405b0bbb91edca3bcef8cbSelective chemical protein modificationSpicer, Christopher D.; Davis, Benjamin G.Nature Communications (2014), 5 (), 4740CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)The chem. modification of proteins is an important tool for probing natural systems and synthesizing novel conjugates. Here, Spicer and Davis review the merits and limitations of the most useful methods for selective modification at both natural and unnatural amino acids.
- 4Zhang, Z., Smith, B. A., Wang, L., Brock, A., Cho, C., and Schultz, P. G. (2003) A new strategy for the site-specific modification of proteins in vivo Biochemistry 42, 6735– 46 DOI: 10.1021/bi03002314https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXjs12qtr8%253D&md5=07c3108f4ce3e4a107c45f6f70db9bb1A New Strategy for the Site-Specific Modification of Proteins in VivoZhang, Zhiwen; Smith, Brian A. C.; Wang, Lei; Brock, Ansgar; Cho, Charles; Schultz, Peter G.Biochemistry (2003), 42 (22), 6735-6746CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)We recently developed a method for genetically incorporating unnatural amino acids site-specifically into proteins expressed in Escherichia coli in response to the amber nonsense codon. Here we describe the selection of an orthogonal tRNA-TyrRS pair that selectively and efficiently incorporates m-acetyl-L-phenylalanine into proteins in E. coli. We demonstrate that proteins contg. m-acetyl-L-phenylalanine or p-acetyl-L-phenylalanine can be selectively labeled with hydrazide derivs. not only in vitro but also in living cells. The labeling reactions are selective and in general proceed with yields of >75%. In specific examples, m-acetyl-L-phenylalanine was substituted for Lys7 of the cytoplasmic protein Z domain, and for Arg200 of the outer membrane protein LamB, and the mutant proteins were selectively labeled with a series of fluorescent dyes. The genetic incorporation of a nonproteinogenic "ketone handle" into proteins provides a powerful tool for the introduction of biophys. probes for the structural and functional anal. of proteins in vitro or in vivo.
- 5Umeda, A., Thibodeaux, G. N., Zhu, J., Lee, Y., and Zhang, Z. J. (2009) Site-specific protein cross-linking with genetically incorporated 3,4-dihydroxy-L-phenylalanine ChemBioChem 10, 1302– 04 DOI: 10.1002/cbic.200900127There is no corresponding record for this reference.
- 6Burdine, L., Gillette, T. G., Lin, H. J., and Kodadek, T. (2004) Periodate-triggered cross-linking of DOPA-containing peptide-protein complexes J. Am. Chem. Soc. 126, 11442– 3 DOI: 10.1021/ja045982cThere is no corresponding record for this reference.
- 7Mao, H., Hart, S. A., Schink, A., and Pollok, B. A. (2004) Sortase-mediated protein ligation: a new method for protein engineering J. Am. Chem. Soc. 126, 2670– 1 DOI: 10.1021/ja039915e7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhtVGisrs%253D&md5=7ae77a5a866ce5e2544550754f086560Sortase-Mediated Protein Ligation: A New Method for Protein EngineeringMao, Hongyuan; Hart, Scott A.; Schink, Amy; Pollok, Brian A.Journal of the American Chemical Society (2004), 126 (9), 2670-2671CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Sortase (SrtA), a transpeptidase from Staphylococcus aureus, catalyzes a cell-wall sorting reaction at an LPXTG motif by cleaving between threonine and glycine and subsequently joining the carboxyl group of threonine to an amino group of pentaglycine on the cell wall peptidoglycan. We have applied this transpeptidyl activity of sortase to in vitro protein ligation. We found that in the presence of sortase, protein/peptide with an LPXTG motif can be specifically ligated to an aminoglycine protein/peptide via an amide bond. Addnl., sortase can even conjugate substrates such as (D)-peptides, synthetic branched peptides, and aminoglycine-derivatized small mols. to the C terminus of a recombinant protein. The sortase-mediate protein ligation is robust, specific, and easy to perform, and can be widely applied to specific protein conjugation with polypeptides or mols. of unique biochem. and biophys. properties.
- 8Schumacher, D., Helma, J., Mann, F. A., Pichler, G., Natale, F., Krause, E., Cardoso, M. C., Hackenberger, C. P. R., and Leonhardt, H. (2015) Versatile and efficient site-specific protein functionalization by tubulin tyrosine ligase Angew. Chem., Int. Ed. 54, 13787– 91 DOI: 10.1002/anie.2015054568https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFGkt7%252FL&md5=5801b9d620c4656b8ce79e6d96d06e72Versatile and Efficient Site-Specific Protein Functionalization by Tubulin Tyrosine LigaseSchumacher, Dominik; Helma, Jonas; Mann, Florian A.; Pichler, Garwin; Natale, Francesco; Krause, Eberhard; Cardoso, M. Cristina; Hackenberger, Christian P. R.; Leonhardt, HeinrichAngewandte Chemie, International Edition (2015), 54 (46), 13787-13791CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)A novel chemoenzymic approach for simple and fast site-specific protein labeling is reported. Recombinant tubulin tyrosine ligase (TTL) was repurposed to attach various unnatural tyrosine derivs. as small bioorthogonal handles to proteins contg. a short tubulin-derived recognition sequence (Tub-tag). This novel strategy enables a broad range of high-yielding and fast chemoselective C-terminal protein modifications on isolated proteins or in cell lysates for applications in biochem., cell biol., and beyond, as demonstrated by the site-specific labeling of nanobodies, GFP, and ubiquitin.
- 9Holder, P. G., Jones, L. C., Drake, P. M., Barfield, R. M., Banas, S., de Hart, G. W., Baker, J., and Rabuka, D. (2015) Reconstitution of formylglycine-generating enzyme with copper(II) for aldehyde tag conversion J. Biol. Chem. 290, 15730– 45 DOI: 10.1074/jbc.M115.6526699https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtVKhsLvN&md5=580235fe160698373c76b21ce354d61bReconstitution of Formylglycine-generating Enzyme with Copper(II) for Aldehyde Tag ConversionHolder, Patrick G.; Jones, Lesley C.; Drake, Penelope M.; Barfield, Robyn M.; Banas, Stefanie; de Hart, Gregory W.; Baker, Jeanne; Rabuka, DavidJournal of Biological Chemistry (2015), 290 (25), 15730-15745CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)To further our aim of synthesizing aldehyde-tagged proteins for research and biotechnol. applications, we developed methods for recombinant prodn. of aerobic formylglycine-generating enzyme (FGE) in good yield. We then optimized the FGE biocatalytic reaction conditions for conversion of cysteine to formylglycine in aldehyde tags on intact monoclonal antibodies. During the development of these conditions, we discovered that pretreating FGE with copper(II) is required for high turnover rates and yields. After further investigation, we confirmed that both aerobic prokaryotic (Streptomyces coelicolor) and eukaryotic (Homo sapiens) FGEs contain a copper cofactor. The complete kinetic parameters for both forms of FGE are described, along with a proposed mechanism for FGE catalysis that accounts for the copper-dependent activity.
- 10Zhang, H., Trout, W. S., Liu, S., Andrade, G. A., Hudson, D. A., Scinto, S. L., Dicker, K. T., Li, Y., Lazouski, N., and Rosenthal, J. 2016, Rapid Bioorthogonal Chemistry Turn-on through Enzymatic or Long Wavelength Photocatalytic Activation of Tetrazine Ligation J. Am. Chem. Soc. 138, 5978– 5983 DOI: 10.1021/jacs.6b0216810https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmtFSntrw%253D&md5=701ccea88f61e6eaf883d240e1785833Rapid Bioorthogonal Chemistry Turn-on through Enzymatic or Long Wavelength Photocatalytic Activation of Tetrazine LigationZhang, Han; Trout, William S.; Liu, Shuang; Andrade, Gabriel A.; Hudson, Devin A.; Scinto, Samuel L.; Dicker, Kevin T.; Li, Yi; Lazouski, Nikifar; Rosenthal, Joel; Thorpe, Colin; Jia, Xinqiao; Fox, Joseph M.Journal of the American Chemical Society (2016), 138 (18), 5978-5983CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Rapid bioorthogonal reactivity can be induced by controllable, catalytic stimuli using air as the oxidant. Methylene blue (4 μM) irradiated with red light (660 nm) catalyzes the rapid oxidn. of a dihydrotetrazine to a tetrazine thereby turning on reactivity toward trans-cyclooctene dienophiles. Alternately, the aerial oxidn. of dihydrotetrazines can be efficiently catalyzed by nanomolar levels of horseradish peroxidase under peroxide-free conditions. Selection of dihydrotetrazine/tetrazine pairs of sufficient kinetic stability in aerobic aq. solns. is key to the success of these approaches. In this work, polymer fibers carrying latent dihydrotetrazines were catalytically activated and covalently modified by trans-cyclooctene conjugates of small mols., peptides, and proteins. In addn. to visualization with fluorophores, fibers conjugated to a cell adhesive peptide exhibited a dramatically increased ability to mediate contact guidance of cells.
- 11McGaughey, G. B., Gagne, M., and Rappe, A. K. (1998) pi-Stacking interactions. Alive and well in proteins J. Biol. Chem. 273, 15458– 15463 DOI: 10.1074/jbc.273.25.1545811https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXktVSmtLw%253D&md5=ffa442b0a21dd4df1199900542428332π-stacking interactions. Alive and well in proteinsMcgaughey, Georgia B.; Gagne, Marc; Rappe, Anthony K.Journal of Biological Chemistry (1998), 273 (25), 15458-15463CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)A representative set of high resoln. x-ray crystal structures of nonhomologous proteins was examd. to det. the preferred positions and orientations of noncovalent interactions between the arom. side-chains of the amino acids, Phe, Tyr, His, and Trp. To study the primary interactions between arom. amino acids, care was taken to examine only isolated pairs (dimers) of amino acids because trimers and higher order clusters of arom. amino acids behave differently than their dimer counterparts. It was found that pairs (dimers) of arom. side-chain amino acids preferentially aligned their resp. arom. rings in an off-centered parallel orientation. Further, it was found that this parallel-displaced structure was 0.5-0.75 kcal/mol more stable than a T-shaped structure for Phe interactions and 1 kcal/mol more stable than a T-shaped structure for the full set of arom. side-chain amino acids. This exptl. detd. structure and energy difference was consistent with ab initio and mol. mechanics calcns. of the benzene dimer; however, the results were not in agreement with previously published analyses of arom. amino acids in proteins. The preferred orientation was referred to as parallel displaced π-stacking.
- 12Struck, A. W., Bennett, M. R., Shepherd, S. A., Law, B. J., Zhuo, Y., Wong, L. S., and Micklefield, J. (2016) An enzyme cascade for selective modification of tyrosine residues in structurally diverse peptides and proteins J. Am. Chem. Soc. 138, 3038– 45 DOI: 10.1021/jacs.5b1092812https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XisVWmu7g%253D&md5=cc3ae0bb185682b8f997b844b0e2dbbcAn Enzyme Cascade for Selective Modification of Tyrosine Residues in Structurally Diverse Peptides and ProteinsStruck, Anna-Winona; Bennett, Matthew R.; Shepherd, Sarah A.; Law, Brian J. C.; Zhuo, Ying; Wong, Lu Shin; Micklefield, JasonJournal of the American Chemical Society (2016), 138 (9), 3038-3045CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Bioorthogonal chem. enables a specific moiety in a complex biomol. to be selectively modified in the presence of many reactive functional groups and other cellular entities. Such selectivity has become indispensable in biol., enabling biomols. to be derivatized, conjugated, labeled, or immobilized for imaging, biochem. assays, or therapeutic applications. Methyltransferase enzymes (MTase) that accept analogs of the cofactor S-adenosyl methionine have been widely deployed for alkyl-diversification and bioorthogonal labeling. However, MTases typically possess tight substrate specificity. Here we introduce a more flexible methodol. for selective derivatization of phenolic moieties in complex biomols. Our approach relies on the tandem enzymic reaction of a fungal tyrosinase and the mammalian catechol-O-methyltransferase (COMT), which can effect the sequential hydroxylation of the phenolic group to give an intermediate catechol moiety that is subsequently O-alkylated. When used in this combination, the alkoxylation is highly selective for tyrosine residues in peptides and proteins, yet remarkably tolerant to changes in the peptide sequence. Tyrosinase-COMT are shown to provide highly versatile and regioselective modification of a diverse range of substrates including peptide antitumor agents, hormones, cyclic peptide antibiotics, and model proteins.
- 13Schlick, T. L., Ding, Z., Kovacs, E. W., and Francis, M. B. (2005) Dual-surface modification of the tobacco mosaic virus J. Am. Chem. Soc. 127, 3718– 23 DOI: 10.1021/ja046239n13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhs1ant7w%253D&md5=417dfb81edc43b89ef121f8c67ed653aDual-surface modification of the Tobacco mosaic virusSchlick, Tara L.; Ding, Zhebo; Kovacs, Ernest W.; Francis, Matthew B.Journal of the American Chemical Society (2005), 127 (11), 3718-3723CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The protein shell of the tobacco mosaic virus (TMV) provides a robust and practical tubelike scaffold for the prepn. of nanoscale materials. To expand the range of applications for which the capsid can be used, two synthetic strategies have been developed for the attachment of new functionality to either the exterior or the interior surface of the virus. The first of these is accomplished using a highly efficient diazonium coupling/oxime formation sequence, which installs >2000 copies of a material component on the capsid exterior. Alternatively, the inner cavity of the tube can be modified by attaching amines to glutamic acid side chains through a carbodiimide coupling reaction. Both of these reactions have been demonstrated for a series of substrates, including biotin, chromophores, and crown ethers. Through the attachment of PEG polymers to the capsid exterior, org.-sol. TMV rods have been prepd. Finally, the orthogonality of these reactions has been demonstrated by installing different functional groups on the exterior and interior surfaces of the same capsid assemblies.
- 14Ban, H., Gavrilyuk, J., and Barbas, C. F., 3rd (2010) Tyrosine bioconjugation through aqueous ene-type reactions: a click-like reaction for tyrosine J. Am. Chem. Soc. 132, 1523– 5 DOI: 10.1021/ja909062q14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXkt1Wrug%253D%253D&md5=876ee34e149ceed4f70f7fd4efbb25baTyrosine bioconjugation through aqueous ene-type reactions: A click-like reaction for tyrosineBan, Hitoshi; Gavrilyuk, Julia; Barbas, Carlos F.Journal of the American Chemical Society (2010), 132 (5), 1523-1525CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)A new and versatile class of cyclic diazodicarboxamides that reacts efficiently and selectively with phenols and the phenolic side chain of tyrosine through an ene-like reaction is reported. This mild aq. tyrosine ligation reaction works over a broad pH range and expands the repertoire of aq. chemistries available for small mol., peptide, and protein modification. The tyrosine ligation reactions are shown to be compatible with the labeling of native enzymes and antibodies in a buffered aq. soln. This reaction provides a novel synthetic approach to bispecific antibodies. The authors believe this reaction will find broad utility in peptide and protein chem. and in the chem. of phenol-contg. compds.
- 15Ban, H., Nagano, M., Gavrilyuk, J., Hakamata, W., Inokuma, T., and Barbas, C. F., 3rd (2013) Facile and stabile linkages through tyrosine: bioconjugation strategies with the tyrosine-click reaction Bioconjugate Chem. 24, 520– 32 DOI: 10.1021/bc300665t15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXkslWlsrY%253D&md5=7f548b32c1e16dd72adbe15b05bbf38cFacile and Stabile Linkages through Tyrosine: Bioconjugation Strategies with the Tyrosine-Click ReactionBan, Hitoshi; Nagano, Masanobu; Gavrilyuk, Julia; Hakamata, Wataru; Inokuma, Tsubasa; Barbas, Carlos F., IIIBioconjugate Chemistry (2013), 24 (4), 520-532CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)The scope, chemoselectivity, and utility of the click-like tyrosine labeling reaction with 4-phenyl-3H-1,2,4-triazoline-3,5(4H)-diones (PTADs) is reported. To study the utility and chemoselectivity of PTAD derivs. in peptide and protein chem., we synthesized PTAD derivs. possessing azide, alkyne, and ketone groups and studied their reactions with amino acid derivs. and peptides of increasing complexity. With proteins we studied the compatibility of the tyrosine click reaction with cysteine and lysine-targeted labeling approaches and demonstrate that chemoselective trifunctionalization of proteins is readily achieved. In particular cases, we noted that PTAD decompn. resulted in formation of a putative isocyanate byproduct that was promiscuous in labeling. This side reaction product, however, was readily scavenged by the addn. of a small amt. of 2-amino-2-hydroxymethyl-propane-1,3-diol (Tris) to the reaction medium. To study the potential of the tyrosine click reaction to introduce poly(ethylene glycol) chains onto proteins (PEGylation), we demonstrate that this novel reagent provides for the selective PEGylation of chymotrypsinogen, whereas traditional succinimide-based PEGylation targeting lysine residues provided a more diverse range of PEGylated products. Finally, we applied the tyrosine click reaction to create a novel antibody-drug conjugate. For this purpose, we synthesized a PTAD deriv. linked to the HIV entry inhibitor aplaviroc. Labeling of the antibody trastuzumab with this reagent provided a labeled antibody conjugate that demonstrated potent HIV-1 neutralization activity demonstrating the potential of this reaction in creating protein conjugates with small mols. The tyrosine click linkage demonstrated stability to extremes of pH, temp., and exposure to human blood plasma indicating that this linkage is significantly more robust than maleimide-type linkages that are commonly employed in bioconjugations. These studies support the broad utility of this reaction in the chemoselective modification of small mols., peptides, and proteins under mild aq. conditions over a broad pH range using a wide variety of biol. acceptable buffers such as phosphate buffered saline (PBS) and 2-amino-2-hydroxymethyl-propane-1,3-diol (Tris) buffers as well as others and mixed buffered compns.
- 16Minamihata, K., Goto, M., and Kamiya, N. (2011) Site-specific protein cross-linking by peroxidase-catalyzed activation of a tyrosine-containing peptide tag Bioconjugate Chem. 22, 74– 81 DOI: 10.1021/bc100398216https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsFGhs7nM&md5=ddc68d94c1e48f91313aa366eefc50e9Site-Specific Protein Cross-Linking by Peroxidase-Catalyzed Activation of a Tyrosine-Containing Peptide TagMinamihata, Kosuke; Goto, Masahiro; Kamiya, NorihoBioconjugate Chemistry (2011), 22 (1), 74-81CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Protein modification methods represent fundamental techniques that are applicable in many fields. In this study, a site-specific protein crosslinking based on the oxidative tyrosine coupling reaction was demonstrated. In the presence of horseradish peroxidase (HRP) and H2O2, tyrosine residues undergo one-electron oxidn. reactions and form radicals in their phenolic moieties, and these species subsequently react with each other to form dimers or further react to generate polymers. Here, a peptide-tag contg. a tyrosine residue(s) (Y-tag, of which the amino acid sequences were either GGGGY or GGYYY) was genetically introduced at the C-terminus of a model protein, Escherichia coli alk. phosphatase (BAP). Following the incubation of recombinant BAPs with HRP and H2O2, Y-tagged BAPs were efficiently cross-linked with each other, whereas wild-type BAP did not undergo crosslinking, indicating that the tyrosine residues in the Y-tags were recognized by HRP as the substrates. To det. the site-specificity of the crosslinking reaction, the Y-tag was selectively removed by thrombin digestion. The resultant BAP without the Y-tag showed no reactivity in the presence of HRP and H2O2. Conversely, Y-tagged BAPs cross-linked by HRP treatment were almost completely digested into monomeric BAP units following incubation with the protease. Moreover, cross-linked Y-tagged BAPs retained ∼95% of their native enzymic activity. These results show that HRP catalyzed the site-specific crosslinking of BAPs through tyrosine residues positioned in the C-terminal Y-tag. The site-selective enzymic oxidative tyrosine coupling reaction should offer a practical option for site-specific and covalent protein modifications.
- 17Tilley, S. D. and Francis, M. B. (2006) Tyrosine-selective protein alkylation using pi-allylpalladium complexes J. Am. Chem. Soc. 128, 1080– 81 DOI: 10.1021/ja057106k17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XitVymsw%253D%253D&md5=8881b14279e96462847e868e6ede7c7eTyrosine-Selective Protein Alkylation Using π-Allylpalladium ComplexesTilley, S. David; Francis, Matthew B.Journal of the American Chemical Society (2006), 128 (4), 1080-1081CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)A new protein modification reaction has been developed based on a palladium-catalyzed allylic alkylation of tyrosine residues. This technique employs electrophilic π-allyl intermediates derived from allylic acetate and carbamate precursors and can be used to modify proteins in aq. soln. at room temp. To facilitate the detection of modified proteins using SDS-PAGE anal., a fluorescent allyl acetate was synthesized and coupled to chymotrypsinogen A and bacteriophage MS2. The tyrosine selectivity of the reaction was confirmed through trypsin digest anal. The utility of the reaction was demonstrated by using taurine-derived carbamates as water solubilizing groups that are cleaved upon protein functionalization. This soly. switching technique was used to install hydrophobic farnesyl and C17 chains on chymotrypsinogen A in water using little or no cosolvent. Following this, the C17 alkylated proteins were found to assoc. with lipid vesicles. In addn. to providing a new protein modification strategy targeting an underutilized amino acid side chain, this method provides convenient access to synthetic lipoproteins.
- 18Romanini, D. W. and Francis, M. B. (2008) Attachment of peptide building blocks to proteins through tyrosine bioconjugation Bioconjugate Chem. 19, 153– 7 DOI: 10.1021/bc700231v18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhsVWgtbbF&md5=e66a855c3a122ae3ea53db9e7d0c9be3Attachment of Peptide Building Blocks to Proteins Through Tyrosine BioconjugationRomanini, Dante W.; Francis, Matthew B.Bioconjugate Chemistry (2008), 19 (1), 153-157CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Recent efforts have yielded a no. of short peptide sequences with useful binding, sensing, and cellular uptake properties. To attach these sequences to tyrosine residues on intact proteins, a three-component Mannich-type strategy is reported. Two solid phase synthetic routes were developed to access peptides up to 20 residues in length with anilines at either the N- or C-termini. In the presence of 20 mM formaldehyde, these functional groups were coupled to tyrosine residues on proteins under mild reaction conditions. The identities of the resulting bioconjugates were confirmed using mass spectrometry and immunoblot anal. Screening expts. have demonstrated that the method is compatible with substrates contg. all of the amino acids, including lysine and cysteine residues. Importantly, tyrosine residues on proteins exhibit much faster reaction rates, allowing short peptides contg. this residue to be coupled without cross reactions.
- 19Long, M. J. C. and Hedstrom, L. (2012) Mushroom tyrosinase oxidizes tyrosine-rich sequences to allow selective protein functionalization ChemBioChem 13, 1818– 25 DOI: 10.1002/cbic.20110079219https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtVCjtL7O&md5=0ad720d91299fcbef8304da0cee16763Mushroom Tyrosinase Oxidizes Tyrosine-Rich Sequences to Allow Selective Protein FunctionalizationLong, Marcus J. C.; Hedstrom, LizbethChemBioChem (2012), 13 (12), 1818-1825, S1818/1-S1818/12CODEN: CBCHFX; ISSN:1439-4227. (Wiley-VCH Verlag GmbH & Co. KGaA)We show that mushroom tyrosinase catalyzes the formation of reactive o-quinones on unstructured, tyrosine-rich sequences such as hemagglutinin (HA) tags (YPYDVPDYA). In the absence of exogenous nucleophiles and at low protein concns., the o-quinone decomps. with fragmentation of the HA tag. At higher protein concns. (>5 mg mL-1), crosslinking is obsd. Besthorn's reagent intercepts the o-quinone to give a characteristic pink complex that can be obsd. directly on a denaturing SDS-PAGE gel. Similar labeled species can be formed by using other nucleophiles such as Cy5-hydrazide. These reactions are selective for proteins bearing HA and other unstructured poly-tyrosine-contg. tags and can be performed in lysates to create specifically tagged proteins.
- 20Faccio, G., Kampf, M. M., Piatti, C., Thony-Meyer, L., and Richter, M. (2014) Tyrosinase-catalyzed site-specific immobilization of engineered C-phycocyanin to surface Sci. Rep. 4, 5370 DOI: 10.1038/srep0537020https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXktlOjt7s%253D&md5=0e61c1970459d54a9709f6a30c0ea080Tyrosinase-catalyzed site-specific immobilization of engineered C-phycocyanin to surfaceFaccio, Greta; Kampf, Michael M.; Piatti, Chiara; Thony-Meyer, Linda; Richter, MichaelScientific Reports (2014), 4 (), 5370CODEN: SRCEC3; ISSN:2045-2322. (Nature Publishing Group)Enzymic crosslinking of proteins is often limited by the steric availability of the target residues, as of tyrosyl side chains in the case of tyrosinase. Carrying an N-terminal peptide-tag contg. two tyrosine residues, the fluorescent protein C-phycocyanin HisCPC from Synechocystis sp. PCC6803 was crosslinked to fluorescent high-mol. wt. forms with tyrosinase. Crosslinking with tyrosinase in the presence of L-tyrosine produced non fluorescent high-mol. wt. products. Incubated in the presence of tyrosinase, HisCPC could also be immobilized to amino-modified polystyrene beads thus conferring a blue fluorescence. Crosslinking and immobilization were site-specific as both processes required the presence of the N-terminal peptide in HisCPC.
- 21Ito, S., Kato, T., Shinpo, K., and Fujita, K. (1984) Oxidation of tyrosine residues in proteins by tyrosinase. Formation of protein-bonded 3,4-dihydroxyphenylalanine and 5-S-cysteinyl-3,4-dihydroxyphenylalanine Biochem. J. 222, 407– 11 DOI: 10.1042/bj222040721https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL2cXlsVOhtLo%253D&md5=c4d30cd7e9a3a9e217ceb2b17cf8d21dOxidation of tyrosine residues in proteins by tyrosinase. Formation of protein-bonded 3,4-dihydroxyphenylalanine and 5-S-cysteinyl-3,4-dihydroxyphenylalanineIto, Shosuke; Kato, Toshiaki; Shinpo, Kan; Fujita, KeisukeBiochemical Journal (1984), 222 (2), 407-11CODEN: BIJOAK; ISSN:0264-6021.A simple and rapid method was developed for the detn. of 3,4-dihydroxyphenylalanine (DOPA) and 5-S-cysteinyl-3,4-dihydroxyphenylalanine (5-S-cysteinyl-DOPA) in proteins with the use of high-pressure liq. chromatog. With this method, it was demonstrated that mushroom tyrosinase can catalyze hydroxylation of tyrosine residues in proteins to DOPA and subsequent oxidn. to dopaquinone residues. The dopaquinone residues in proteins combine with cysteine residues to form 5-S-cysteinyl-DOPA in bovine serum albumin and yeast alc. dehydrogenase, whereas DOPA is the major product in bovine insulin which lacks cysteine residues.
- 22Tabakovic, K. and Abul-Hajj, Y. J. (1994) Reaction of lysine with estrone 3,4-o-quinone Chem. Res. Toxicol. 7, 696– 701 DOI: 10.1021/tx00041a016There is no corresponding record for this reference.
- 23Xu, R., Huang, X., Morgan, T. D., Prakash, O., Kramer, K. J., and Hawley, M. D. (1996) Characterization of products from the reactions of N-acetyldopamine quinone with N-acetylhistidine Arch. Biochem. Biophys. 329, 56– 64 DOI: 10.1006/abbi.1996.0191There is no corresponding record for this reference.
- 24Borrmann, A., Fatunsin, O., Dommerholt, J., Jonker, A. M., Lowik, D. W., van Hest, J. C., and van Delft, F. L. (2015) Strain-promoted oxidation-controlled cyclooctyne-1,2-quinone cycloaddition (SPOCQ) for fast and activatable protein conjugation Bioconjugate Chem. 26, 257– 61 DOI: 10.1021/bc500534d24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitFCrs7vK&md5=7b072746ff08db93e05ad0dac3ae0966Strain-Promoted Oxidation-Controlled Cyclooctyne-1,2-Quinone Cycloaddition (SPOCQ) for Fast and Activatable Protein ConjugationBorrmann, Annika; Fatunsin, Olumide; Dommerholt, Jan; Jonker, Anika M.; Loewik, Dennis W. P. M.; van Hest, Jan C. M.; van Delft, Floris L.Bioconjugate Chemistry (2015), 26 (2), 257-261CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)A main challenge in the area of bioconjugation is to devise reactions that are both activatable and fast. Here, we introduce a temporally controlled reaction between cyclooctynes and 1,2-quinones, induced by facile oxidn. of 1,2-catechols. This so-called strain-promoted oxidn.-controlled cyclooctyne-1,2-quinone cycloaddn. (SPOCQ) shows a remarkably high reaction rate when performed with bicyclononyne (BCN), out-competing the well-known cycloaddn. of azides and BCN by 3 orders of magnitude, thereby allowing a new level of orthogonality in protein conjugation.
- 25Jonker, A. M., Borrmann, A., van Eck, E. R., van Delft, F. L., Lowik, D. W., and van Hest, J. C. (2015) A fast and activatable cross-linking strategy for hydrogel formation Adv. Mater. 27, 1235– 40 DOI: 10.1002/adma.20140444825https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitFamsbbI&md5=c6d61b6bfee10fb1566884006b36fd1cA Fast and Activatable Cross-Linking Strategy for Hydrogel FormationJonker, Anika M.; Borrmann, Annika; van Eck, Ernst R. H.; van Delft, Floris L.; Loewik, Dennis W. P. M.; van Hest, Jan C. M.Advanced Materials (Weinheim, Germany) (2015), 27 (7), 1235-1240CODEN: ADVMEW; ISSN:0935-9648. (Wiley-VCH Verlag GmbH & Co. KGaA)The study reports a fast and activatible crosslinking method for hydrogel formation using a a strain-promoted oxidn. controlled cyclooctyne-1,2-quinone cycloaddn.
- 26Dommerholt, J., Schmidt, S., Temming, R., Hendriks, L. J., Rutjes, F. P., van Hest, J. C., Lefeber, D. J., Friedl, P., and van Delft, F. L. (2010) Readily accessible bicyclononynes for bioorthogonal labeling and three-dimensional imaging of living cells Angew. Chem., Int. Ed. 49, 9422– 5 DOI: 10.1002/anie.20100376126https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsFSls73M&md5=1d18345acd06c8b7042111caccfdca06Readily accessible bicyclononynes for bioorthogonal labeling and three-dimensional imaging of living cellsDommerholt, Jan; Schmidt, Samuel; Temming, Rinske; Hendriks, Linda J. A.; Rutjes, Floris P. J. T.; van Hest, Jan C. M.; Lefeber, Dirk J.; Friedl, Peter; van Delft, Floris L.Angewandte Chemie, International Edition (2010), 49 (49), 9422-9425, S9422/1-S9422/22CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)We reported to use bicyclo[6.1.0]nonyne (BCN) as a novel ring-strained alkyne for metal-free cycloaddn. reactions with azides and nitrones. The complexes were synthesized and used for bioorthogonal labeling/three-dimensional imaging of living cells.
- 27Przybysz, A., Volmer, A. A., Westphal, A. H., and van Berkel, W. J. (2014) Bifunctional immobilization of a hyperthermostable endo-beta-1,3-glucanase Appl. Microbiol. Biotechnol. 98, 1155– 63 DOI: 10.1007/s00253-013-4953-3There is no corresponding record for this reference.
- 28Wagner, K., Kwakkenbos, M. J., Claassen, Y. B., Maijoor, K., Bohne, M., van der Sluijs, K. F., Witte, M. D., van Zoelen, D. J., Cornelissen, L. A., and Beaumont, T. 2014, Bispecific antibody generated with sortase and click chemistry has broad antiinfluenza virus activity Proc. Natl. Acad. Sci. U. S. A. 111, 16820– 5 DOI: 10.1073/pnas.140860511128https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvV2gsLvN&md5=51a3e79489b81bc279bd38177c1c4d94Bispecific antibody generated with sortase and click chemistry has broad antiinfluenza virus activityWagner, Koen; Kwakkenbos, Mark J.; Claassen, Yvonne B.; Maijoor, Kelly; Boehne, Martino; van der Sluijs, Koenraad F.; Witte, Martin D.; van Zoelen, Diana J.; Cornelissen, Lisette A.; Beaumont, Tim; Bakker, Arjen Q.; Ploegh, Hidde L.; Spits, HergenProceedings of the National Academy of Sciences of the United States of America (2014), 111 (47), 16820-16825CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Bispecific antibodies have therapeutic potential by expanding the functions of conventional antibodies. Many different formats of bispecific antibodies have meanwhile been developed. Most are genetic modifications of the antibody backbone to facilitate incorporation of two different variable domains into a single mol. Here, we present a bispecific format where we have fused two full-sized IgG antibodies via their C termini using sortase trans-peptidation and click chem. to create a covalently linked IgG antibody heterodimer. By linking two potent anti-influenza A antibodies together, we have generated a full antibody dimer with bispecific activity that retains the activity and stability of the two fusion partners.
- 29Dorywalska, M., Strop, P., Melton-Witt, J. A., Hasa-Moreno, A., Farias, S. E., Galindo Casas, M., Delaria, K., Lui, V., Poulsen, K., and Loo, C. 2015, Effect of Attachment Site on Stability of Cleavable Antibody Drug Conjugates Bioconjugate Chem. 26, 650– 659 DOI: 10.1021/bc500574729https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvFGnsb4%253D&md5=c6ea72edbc33b0ad4aa743bc21707401Effect of Attachment Site on Stability of Cleavable Antibody Drug ConjugatesDorywalska, Magdalena; Strop, Pavel; Melton-Witt, Jody A.; Hasa-Moreno, Adela; Farias, Santiago E.; Galindo Casas, Meritxell; Delaria, Kathy; Lui, Victor; Poulsen, Kris; Loo, Carole; Krimm, Stellanie; Bolton, Gary; Moine, Ludivine; Dushin, Russell; Tran, Thomas-Toan; Liu, Shu-Hui; Rickert, Mathias; Foletti, Davide; Shelton, David L.; Pons, Jaume; Rajpal, ArvindBioconjugate Chemistry (2015), 26 (4), 650-659CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)The systemic stability of the antibody-drug linker is crucial for delivery of an intact antibody-drug conjugate (ADC) to target-expressing tumors. Linkers stable in circulation but readily processed in the target cell are necessary for both safety and potency of the delivered conjugate. Here, we report a range of stabilities for an auristatin-based payload site-specifically attached through a cleavable valine-citrulline-p-aminobenzylcarbamate (VC-PABC) linker across various sites on an antibody. We demonstrate that the conjugation site plays an important role in detg. VC-PABC linker stability in mouse plasma, and that the stability of the linker pos. correlates with ADC cytotoxic potency both in vitro and in vivo. Furthermore, we show that the VC-PABC cleavage in mouse plasma is not mediated by Cathepsin B, the protease thought to be primarily responsible for linker processing in the lysosomal degrdn. pathway. Although the VC-PABC cleavage is not detected in primate plasma in vitro, linker stabilization in the mouse is an essential prerequisite for designing successful efficacy and safety studies in rodents during preclin. stages of ADC programs. The divergence of linker metab. in mouse plasma and its intracellular cleavage offers an opportunity for linker optimization in the circulation without compromising its efficient payload release in the target cell.
- 30Chari, R. V., Miller, M. L., and Widdison, W. C. (2014) Antibody-drug conjugates: an emerging concept in cancer therapy Angew. Chem., Int. Ed. 53, 3796– 827 DOI: 10.1002/anie.20130762830https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXjtVGmsr0%253D&md5=fbfb2f843f516654ea7c0eb98949604cAntibody-Drug Conjugates: An Emerging Concept in Cancer TherapyChari, Ravi V. J.; Miller, Michael L.; Widdison, Wayne C.Angewandte Chemie, International Edition (2014), 53 (15), 3796-3827CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Traditional cancer chemotherapy is often accompanied by systemic toxicity to the patient. Monoclonal antibodies against antigens on cancer cells offer an alternative tumor-selective treatment approach. However, most monoclonal antibodies are not sufficiently potent to be therapeutically active on their own. Antibody-drug conjugates (ADCs) use antibodies to deliver a potent cytotoxic compd. selectively to tumor cells, thus improving the therapeutic index of chemotherapeutic agents. The recent approval of two ADCs, brentuximab vedotin and ado-trastuzumab emtansine, for cancer treatment has spurred tremendous research interest in this field. This Review touches upon the early efforts in the field, and describes how the lessons learned from the first-generation ADCs have led to improvements in every aspect of this technol., i.e., the antibody, the cytotoxic compd., and the linker connecting them, leading to the current successes. The design of ADCs currently in clin. development, and results from mechanistic studies and preclin. and clin. evaluation are discussed. Emerging technologies that seek to further advance this exciting area of research are also discussed.
- 31Younes, A., Bartlett, N. L., Leonard, J. P., Kennedy, D. A., Lynch, C. M., Sievers, E. L., and Forero-Torres, A. (2010) Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas N. Engl. J. Med. 363, 1812– 21 DOI: 10.1056/NEJMoa100296531https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtl2gsbbE&md5=d27d22f971143a6c5415090ab57fe01aBrentuximab vedotin (SGN-35) for relapsed Cd30-positive lymphomasYounes, Anas; Bartlett, Nancy L.; Leonard, John P.; Kennedy, Dana A.; Lynch, Carmel M.; Sievers, Eric L.; Forero-Torres, AndresNew England Journal of Medicine (2010), 363 (19), 1812-1821CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)BACKGROUND: Hodgkin's lymphoma and anaplastic large-cell lymphoma are the two most common tumors expressing CD30. Previous attempts to target the CD30 antigen with monoclonal-based therapies have shown minimal activity. To enhance the antitumor activity of CD30-directed therapy, the antitubulin agent monomethyl auristatin E (MMAE) was attached to a CD30-specific monoclonal antibody by an enzyme-cleavable linker, producing the antibody-drug conjugate brentuximab vedotin (SGN-35). METHODS: In this phase 1, open-label, multicenter dose-escalation study, we administered brentuximab vedotin (at a dose of 0.1 to 3.6 mg per kg of body wt.) every 3 wk to 45 patients with relapsed or refractory CD30-pos. hematol. Cancers, primarily Hodgkin's lymphoma and anaplastic large-cell lymphoma. Patients had received a median of three previous chemotherapy regimens (range, one to seven), and 73% had undergone autologous stem-cell transplantation. RESULTS: The max. tolerated dose was 1.8 mg per kg, administered every 3 wk. Objective responses, including 11 complete remissions, were obsd. in 17 patients. Of 12 patients who received the 1.8-mg-per-kilogram dose, 6 (50%) had an objective response. The median duration of response was at least 9.7 mo. Tumor regression was obsd. in 36 of 42 patients who could be evaluated (86%). The most common adverse events were fatigue, pyrexia, diarrhea, nausea, neutropenia, and peripheral neuropathy. CONCLUSIONS: Brentuximab vedotin induced durable objective responses and resulted in tumor regression for most patients with relapsed or refractory CD30-pos. lymphomas in this phase 1 study. Treatment was assocd. primarily with grade 1 or 2 (mild-to-moderate) toxic effects.
- 32Verma, S., Miles, D., Gianni, L., Krop, I. E., Welslau, M., Baselga, J., Pegram, M., Oh, D. Y., Dieras, V., and Guardino, E. 2012, Trastuzumab emtansine for HER2-positive advanced breast cancer N. Engl. J. Med. 367, 1783– 91 DOI: 10.1056/NEJMoa120912432https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhs1ekt73M&md5=3be5bac27a81b530442dc8a6b4a1f82eTrastuzumab emtansine for HER2-positive advanced breast cancerVerma, Sunil; Miles, David; Gianni, Luca; Krop, Ian E.; Welslau, Manfred; Baselga, Jose; Pegram, Mark; Oh, Do-Youn; Dieras, Veronique; Guardino, Ellie; Fang, Liang; Lu, Michael W.; Olsen, Steven; Blackwell, KimNew England Journal of Medicine (2012), 367 (19), 1783-1791CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)BACKGROUND: Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate incorporating the human epidermal growth factor receptor 2 (HER2)-targeted antitumor properties of trastuzumab with the cytotoxic activity of the microtubule-inhibitory agent DM1. The antibody and the cytotoxic agent are conjugated by means of a stable linker. METHODS: We randomly assigned patients with HER2-pos. advanced breast cancer, who had previously been treated with trastuzumab and a taxane, to T-DM1 or lapatinib plus capecitabine. The primary end points were progression-free survival (as assessed by independent review), overall survival, and safety. Secondary end points included progression-free survival (investigator-assessed), the objective response rate, and the time to symptom progression. Two interim analyses of overall survival were conducted. RESULTS: Among 991 randomly assigned patients, median progression-free survival as assessed by independent review was 9.6 mo with T-DM1 vs. 6.4 mo with lapatinib plus capecitabine (hazard ratio for progression or death from any cause, 0.65; 95% confidence interval [CI], 0.55 to 0.77; P<0.001), and median overall survival at the second interim anal. crossed the stopping boundary for efficacy (30.9 mo vs. 25.1 mo; hazard ratio for death from any cause, 0.68; 95% CI, 0.55 to 0.85; P<0.001). The objective response rate was higher with T-DM1 (43.6%, vs. 30.8% with lapatinib plus capecitabine; P<0.001); results for all addnl. secondary end points favored T-DM1. Rates of grade 3 or above were higher with lapatinib plus capecitabine than with T-DM1 (57% vs. 41%). The incidences of thrombocytopenia and increased serum aminotransferase levels were higher with T-DM1, whereas the incidences of diarrhea, nausea, vomiting, and palmar-plantar erythrodysesthesia were higher with lapatinib plus capecitabine. CONCLUSIONS: T-DM1 significantly prolonged progression-free and overall survival with less toxicity than lapatinib plus capecitabine in patients with HER2-pos. advanced breast cancer previously treated with trastuzumab and a taxane.
Supporting Information
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.bioconjchem.7b00046.
Additional method and material details, SPOCQ and expression details, gene and protein sequences, a schematic view of the reaction and corresponding mass values, and MS data. (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.