Tyrosinase-Mediated Synthesis of Nanobody–Cell Conjugates
- Johnathan C. MazaJohnathan C. MazaDepartment of Chemistry, University of California, Berkeley, California 94720, United StatesMore by Johnathan C. Maza
- ,
- Derek M. García-AlmedinaDerek M. García-AlmedinaDepartment of Chemistry, University of California, Berkeley, California 94720, United StatesMore by Derek M. García-Almedina
- ,
- Lydia E. BoikeLydia E. BoikeDepartment of Chemistry, University of California, Berkeley, California 94720, United StatesNovartis-Berkeley Center for Proteomics and Chemistry Technologies, Cambridge, Massachusetts 02139, United StatesMore by Lydia E. Boike
- ,
- Noah X. HamlishNoah X. HamlishDepartment of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720, United StatesMore by Noah X. Hamlish
- ,
- Daniel K. NomuraDaniel K. NomuraDepartment of Chemistry, University of California, Berkeley, California 94720, United StatesNovartis-Berkeley Center for Proteomics and Chemistry Technologies, Cambridge, Massachusetts 02139, United StatesDepartment of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720, United StatesDepartment of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, California 94720, United StatesInnovative Genomics Institute, Berkeley, California 94720, United StatesMore by Daniel K. Nomura
- , and
- Matthew B. Francis*Matthew B. Francis*Email: [email protected]Department of Chemistry, University of California, Berkeley, California 94720, United StatesMaterials Sciences Division, Lawrence Berkeley National Laboratories, Berkeley, California 94720,United StatesMore by Matthew B. Francis
Abstract

A convenient enzymatic strategy is reported for the modification of cell surfaces. Using a tyrosinase enzyme isolated from Agaricus bisporus, unique tyrosine residues introduced at the C-termini of nanobodies can be site-selectively oxidized to reactive o-quinones. These reactive intermediates undergo rapid modification with nucleophilic thiol, amine, and imidazole residues present on cell surfaces, producing novel nanobody–cell conjugates that display targeted antigen binding. We extend this approach toward the synthesis of nanobody–NK cell conjugates for targeted immunotherapy applications. The resulting NK cell conjugates exhibit targeted cell binding and elicit targeted cell death.
This publication is licensed under
License Summary*
You are free to share (copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share (copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share (copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share (copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share (copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Synopsis
A novel methodology is presented for using tyrosinase to synthesize protein−cell conjugates while retaining cell viability and function.
Introduction
Figure 1

Figure 1. General strategy for modifying cell surfaces with nanobodies. (a) Tyrosinase catalyzes the oxidation of small-molecule phenols to highly reactive o-quinones, which can modify nucleophiles present on proteins. Engineered tyrosine tags at protein termini can also be oxidized by tyrosinase, producing a site-specific o-quinone on the protein that reacts with protein-based nucleophiles. (b) Tyrosine-tagged nanobodies can be site-specifically oxidized by tyrosinase for attachment of these proteins to cells. The resulting linkage produces a well-defined point of attachment for installing nanobodies on cell surfaces while imbuing the target cell with novel antigen-binding functionality. (c) Nanobodies are low-molecular-weight (∼10–15 kDa) antigen-binders derived from the variable region of the camelid antibody (PDB ID 3K1K).
Results and Discussion
Tyrosinase-Mediated Synthesis of Nanobody–Cell Conjugates
Figure 2

Figure 2. Modification of NK cell surfaces with nanobodies. (a) Tyrosinase enzyme produces a site-specific o-quinone at the C-terminal Ser–Gly4–Tyr tag installed on nanobodies, as evidenced by a 14 Da mass shift detected via ESI-TOF MS. (b) To verify that NK cell surfaces can be decorated with nanobodies using tyorsinase, a Tyr-tagged nanobody against GFP (nbGFPTyr) was designed. Using tyrosinase, nbGFPTyr can be attached to the cell surface, and 2° labeling with GFP can be used to analyze the reaction using flow cytometry. (c) Labeling experiments with nbGFPTyr validated attachment of the nanobody to the cell surface, as only cells treated with both nbGFPTyr and tyrosinase showed an increase in GFP fluorescence (red trace) over controls (blue and orange traces). (d) Using a Cys point mutant, a single FITC dye can be attached to each nbGFPTyr (nbFITC). After attachment of 10 μM nbFITC to cell surfaces, comparison against FITC-calibration beads determined that a median value of ∼120,000 copies of the nanobody were linked to the cells. Data are represented as box plots, with the top of the box representing the 75th percentile of the data, the middle line representing the median of the data, and the bottom of the box representing the 25th percentile of the data.
Targeted Cell–Cell Interactions in abTYR-Synthesized Nanobody–NK Cell Conjugates
Figure 3

Figure 3. Decoration of NK cells for nanobody-directed cell–cell interactions. (a) Using tyrosinase, a Tyr-tagged nanobody against HER2 (nbHER2Tyr) was attached to NK cells. Secondary labeling with a soluble FITC–HER2 showed that only cells exposed to nbHER2Tyr and tyrosinase exhibited a shift in FITC signal detected via flow cytometry (red trace) over controls. (b) To assess if tyrosinase-synthesized NK–nbHER2 conjugates can make targeted contacts with HER2+ cells, NK–nbHER2 cells were mixed with a HER2+ cell line (SK-BR-3) at a ratio of 2:1 (NK:target). Cells were allowed to bind and settle and then imaged using fluorscence microscopy. A nearest neighbor analysis was performed (CellProfiler), indicating that a statistically significant proportion of target cells (green) were bound to two or more NK–nbHER2 cells (red) only when the NK cells were pretreated with nbHER2Tyr and tyrosinase (orange bar). (c) Fluorescence microscopy images confirm rosette formation is only seen when NK cells are pretreated with both nbHER2Tyr and tyrosinase.
Targeted Killing of nbHER2Tyr–NK Cell Conjugates
Figure 4

Figure 4. Targeted cell killing elicited by tyrosinase-synthesized nanobody–NK cell conjugates. (a) Schematic representation of the fluorescence-based cell assay used to determine NK cytotoxicity. HER2+ cells (SK-BR-3) were preloaded with calcein AM dye, which is retained by the cell membrane after uptake. Lysis of the HER2+ cell releases dye into the supernatant, providing a measurement for cell lysis. Only NK cells pretreated with both nbHER2Tyr and tyrosinase (orange bar) show statistically significant specific cell lysis over control treatments. (b) To assess how the ratio of NK:target cell impacts specific cytotoxicity, NK–nbHER2 cells were synthesized using 10 μM nbHER2Tyr and 400 nM tyrosinase and mixed with calcein AM loaded HER2+ cells (SK-BR-3). Statistically significant cell death was observed at ratios even as low as 2:1 (effector:target). (c) To assess the required concentration of nbHER2Tyr needed to elict NK-mediated cell death, a variety of concentrations of nbHER2Tyr were used to label NK cells with tyrosinase. Increased lysis was observed when using 5 and 10 μM nbHER2, while a sharp reduction of NK lytic activity was observed at the higher concentration of 20 μM nbHER2Tyr.
Conclusion
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acscentsci.1c01265.
Raw peptide lists for small-molecule proteomics (XLSX)
Unique peptides for small-molecule proteomics (XLSX)
Full experimental details and cloning procedures (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.
Acknowledgments
This work was supported by the N.I.H. (R01GM138693) as well as the Chemical Biology Graduate Program at UC Berkeley (NIH T32-GM066698). J.C.M. was supported by a UC Berkeley Fellowship for Graduate Studies, and J.C.M. and L.B. were both supported by NSF Graduate Research Program Fellowships. D.M.G.A. was supported by a UC Berkeley Chancellor Fellowship. The authors thank Paul Huang for assistance in confocal imaging. The authors thank Dr. Mary West for flow cytometry and microscopy resources through the QB3 Cell and Tissue Analysis Facility.
abTYR | tyrosinase from Agaricus bisporus |
sfGFP | superfolder green fluorescent protein |
nbGFPTyr | GFP-binding nanobody with C-terminal Ser–Gly4–Tyr tag |
nbGFPCys | GFP-binding nanobody with C-terminal Ser–Gly4–Cys tag |
nbFITC | A76C mutant of nbGFPTyr, modified with FITC-maleimide |
HER2 | Human epidermal growth factor receptor 2 |
nbHER2Tyr | HER2-binding nanobody with C-terminal Ser–Gly4–Tyr tag |
DPBS | Dulbecco’s phosphate buffered saline solution without Ca2+ or Mg2+ |
References
This article references 34 other publications.
- 1El Muslemany, K. M.; Twite, A. A.; ElSohly, A. M.; Obermeyer, A. C.; Mathies, R. A.; Francis, M. B. Photoactivated Bioconjugation Betweenortho-Azidophenols and Anilines: A Facile Approach to Biomolecular Photopatterning. J. Am. Chem. Soc. 2014, 136, 12600– 12606, DOI: 10.1021/ja503056x[ACS Full Text
], [CAS], Google Scholar
1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsVGht7bL&md5=4c5e84d4074baace09004685a2f0b852Photoactivated Bioconjugation Between ortho-Azidophenols and Anilines: A Facile Approach to Biomolecular PhotopatterningEl Muslemany, Kareem M.; Twite, Amy A.; El Sohly, Adel M.; Obermeyer, Allie C.; Mathies, Richard A.; Francis, Matthew B.Journal of the American Chemical Society (2014), 136 (36), 12600-12606CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Methods for the surface patterning of small mols. and biomols. can yield useful platforms for drug screening, synthetic biol. applications, diagnostics, and the immobilization of live cells. However, new techniques are needed to achieve the ease, feature sizes, reliability, and patterning speed necessary for widespread adoption. Herein, the authors report an easily accessible and operationally simple photoinitiated reaction that can achieve patterned bioconjugation in a highly chemoselective manner. The reaction involves the photolysis of 2-azidophenols to generate iminoquinone intermediates that couple rapidly to aniline groups. The authors demonstrate the broad functional group compatibility of this reaction for the modification of proteins, polymers, oligonucleotides, peptides, and small mols. As a specific application, the reaction was adapted for the photolithog. patterning of azidophenol DNA on aniline glass substrates. The presence of the DNA was confirmed by the ability of the surface to capture living cells bearing the sequence complement on their cell walls or cytoplasmic membranes. Compared to other light-based DNA patterning methods, this reaction offers higher speed and does not require the use of a photoresist or other blocking material. - 2Furst, A. L.; Smith, M. J.; Francis, M. B. Direct Electrochemical Bioconjugation on Metal Surfaces. J. Am. Chem. Soc. 2017, 139, 12610– 12616, DOI: 10.1021/jacs.7b06385[ACS Full Text
], [CAS], Google Scholar
2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsVSlu7%252FI&md5=919d942661541624c0a425c5d6d8c3a4Direct Electrochemical Bioconjugation on Metal SurfacesFurst, Ariel L.; Smith, Matthew J.; Francis, Matthew B.Journal of the American Chemical Society (2017), 139 (36), 12610-12616CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)DNA has unique capabilities for mol. recognition and self-assembly, which have fostered its widespread incorporation into devices that are useful in science and medicine. Many of these platforms rely on thiol groups to tether DNA to gold surfaces, but this method is hindered by a lack of control over monolayer d. and by secondary interactions between the nucleotide bases and the metal. In this work, the authors report an electrochem. activated bioconjugation reaction as a mild, reagent-free strategy to attach oligonucleotides to gold surfaces. Aniline-modified DNA was coupled to catechol-coated electrodes that were oxidized to o-quinones using an applied potential. High levels of coupling could be achieved in minutes. By changing the reaction time and the underlying catechol content, the final DNA surface coverage could be specified. The advantages of this method were demonstrated through the electrochem. detection of the endocrine disruptor bisphenol A, as well as the capture of living nonadherent cells on electrode surfaces by DNA hybridization. This method not only improves the attachment of DNA to metal surfaces but also represents a new direction for the site-specific attachment of biomols. to device platforms. - 3Twite, A. A.; Hsiao, S. C.; Onoe, H.; Mathies, R. A.; Francis, M. B. Direct Attachment of Microbial Organisms to Material Surfaces Through Sequence-Specific DNA Hybridization. Adv. Mater. 2012, 24, 2380– 2385, DOI: 10.1002/adma.201104336[Crossref], [PubMed], [CAS], Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XktFShtLg%253D&md5=ea091e8667c10160fd77d77992e25ea6Direct attachment of microbial organisms to material surfaces through sequence-specific DNA hybridizationTwite, Amy A.; Hsiao, Sonny C.; Onoe, Hiroaki; Mathies, Richard A.; Francis, Matthew B.Advanced Materials (Weinheim, Germany) (2012), 24 (18), 2380-2385CODEN: ADVMEW; ISSN:0935-9648. (Wiley-VCH Verlag GmbH & Co. KGaA)General methods are described for the immobilization of microorganisms on patterned surfaces. A new oxidative strategy for the introduction of nucleic acid strands on microbial cell surfaces results in their strong, efficient, and specific binding to surfaces displaying the sequence complement without apparent losses in cell viability. The two-step method to attach ssDNA to these cell types comprises the initial oxidn. of the 1,2-diols in the carbohydrates with sodium periodate, followed by condensation of the resulting aldehyde groups with 5'-hydrazide-terminated ssDNA to form hydrazones. Hydrazone formation was facilitated by the addn. of aniline (10-25 mM) to the reaction mixt. as a catalyst. After optimizing the cell modification scheme for ssDNA, the ability of the strands to mediate surface adhesion was demonstrated with glass slides prepd. with circular patterns of complementary ssDNA using a previously reported reductive animation procedure. Particular advantages of this approach include its high binding efficiency, its minimal impact on cell viability, and its ability to create complex patterns consisting of many different organisms.
- 4Gartner, Z. J.; Bertozzi, C. R. Programmed Assembly of 3-Dimensional Microtissues with Defined Cellular Connectivity. Proc. National Acad. Sci. 2009, 106, 4606– 4610, DOI: 10.1073/pnas.0900717106[Crossref], [PubMed], [CAS], Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXktFOgsLg%253D&md5=00ecc4ecb82b8359ed33a0ff4d418ff6Programmed assembly of 3-dimensional microtissues with defined cellular connectivityGartner, Zev J.; Bertozzi, Carolyn R.Proceedings of the National Academy of Sciences of the United States of America (2009), 106 (12), 4606-4610CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Multicellular organs comprise differentiated cell types with discrete yet interdependent functions. The cells' spatial arrangements and interconnectivities, both crit. elements of higher-order function, derive from complex developmental programs in vivo and are often difficult or impossible to emulate in vitro. Here, we report the bottom-up synthesis of microtissues composed of multiple cell types with programmed connectivity. We functionalized cells with short oligonucleotides to impart specific adhesive properties. Hybridization of complementary DNA sequences enabled the assembly of multicellular structures with defined cell-cell contacts. We demonstrated that the kinetic parameters of the assembly process depend on DNA sequence complexity, d., and total cell concn. Thus, cell assembly can be highly controlled, enabling the design of microtissues with defined cell compn. and stoichiometry. We used this strategy to construct a paracrine signaling network in isolated 3-dimensional microtissues.
- 5Li, D.; Li, X.; Zhou, W.-L.; Huang, Y.; Liang, X.; Jiang, L.; Yang, X.; Sun, J.; Li, Z.; Han, W.-D.; Wang, W. Genetically Engineered T Cells for Cancer Immunotherapy. Signal Transduct. Target Ther. 2019, 4, 35, DOI: 10.1038/s41392-019-0070-9[Crossref], [PubMed], [CAS], Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MjgtlSqsQ%253D%253D&md5=bbfa61151e99ab8538ac41cf89867e40Genetically engineered T cells for cancer immunotherapyLi Dan; Li Xue; Zhou Wei-Lin; Huang Yong; Liang Xiao; Jiang Lin; Yang Xiao; Wang Wei; Liang Xiao; Sun Jie; Sun Jie; Li Zonghai; Li Zonghai; Han Wei-DongSignal transduction and targeted therapy (2019), 4 (), 35 ISSN:.T cells in the immune system protect the human body from infection by pathogens and clear mutant cells through specific recognition by T cell receptors (TCRs). Cancer immunotherapy, by relying on this basic recognition method, boosts the antitumor efficacy of T cells by unleashing the inhibition of immune checkpoints and expands adaptive immunity by facilitating the adoptive transfer of genetically engineered T cells. T cells genetically equipped with chimeric antigen receptors (CARs) or TCRs have shown remarkable effectiveness in treating some hematological malignancies, although the efficacy of engineered T cells in treating solid tumors is far from satisfactory. In this review, we summarize the development of genetically engineered T cells, outline the most recent studies investigating genetically engineered T cells for cancer immunotherapy, and discuss strategies for improving the performance of these T cells in fighting cancers.
- 6Abbina, S.; Siren, E. M. J.; Moon, H.; Kizhakkedathu, J. N. Surface Engineering for Cell-Based Therapies: Techniques for Manipulating Mammalian Cell Surfaces. ACS Biomater. Sci. Eng. 2018, 4, 3658– 3677, DOI: 10.1021/acsbiomaterials.7b00514[ACS Full Text
], [CAS], Google Scholar
6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsVKmtbnN&md5=d38c4c7e8650957daec8026a52084dc3Surface Engineering for Cell-Based Therapies: Techniques for Manipulating Mammalian Cell SurfacesAbbina, Srinivas; Siren, Erika M. J.; Moon, Haisle; Kizhakkedathu, Jayachandran N.ACS Biomaterials Science & Engineering (2018), 4 (11), 3658-3677CODEN: ABSEBA; ISSN:2373-9878. (American Chemical Society)A review. The introduction of cell-based therapies has provided new and unique strategies to treat many diseases and disorders including the recent approval of CAR-T cell therapy for the leukemia. Cell surface engineering is a methodol. in which the cell surface is tailored to modulate cellular function and interactions. In addn. to genetic engineering of cell surface proteins, a wide array of robust, innovative and elegant approaches have been developed to selectivity target the cell surface. In this review, we will introduce the leading strategies currently used in cell surface engineering including broadly reactive chem. ligations and phys. assocns. as well as more controlled approaches as demonstrated in genetic, enzymic and metabolic engineering. Prominent applications of these strategies for cell-based therapies will be highlighted including targeted cell death, controlling stem cell fate, immunoevasion, blood transfusion and the delivery of cells to target tissues. Advances will be focused specifically on cells which are the most promising in generating cell-based therapeutics including red blood cells, white blood cells (lymphocytes, macrophages), stem cells (multipotent and pluripotent), islet cells, cancer cells and endothelial cells. - 7Stephan, M. T.; Irvine, D. J. Enhancing Cell Therapies from the Outside in: Cell Surface Engineering Using Synthetic Nanomaterials. Nano Today. 2011, 6, 309– 325, DOI: 10.1016/j.nantod.2011.04.001[Crossref], [PubMed], [CAS], Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXpvVWqtbc%253D&md5=2a9955bfe0a5a1d915a7d58063464149Enhancing cell therapies from the outside in: cell surface engineering using synthetic nanomaterialsStephan, Matthias T.; Irvine, Darrell J.Nano Today (2011), 6 (3), 309-325CODEN: NTAOCG; ISSN:1748-0132. (Elsevier Ltd.)A review. Therapeutic treatments based on the injection of living cells are in clin. use and preclin. development for diseases ranging from cancer to cardiovascular disease to diabetes. To enhance the function of therapeutic cells, a variety of chem. and materials science strategies are being developed that engineer the surface of therapeutic cells with new mols., artificial receptors, and multifunctional nanomaterials, synthetically endowing donor cells with new properties and functions. These approaches offer a powerful complement to traditional genetic engineering strategies for enhancing the function of living cells.
- 8Shearer, R. F.; Saunders, D. N. Experimental Design for Stable Genetic Manipulation in Mammalian Cell Lines: Lentivirus and Alternatives. Genes Cells. 2015, 20, 1– 10, DOI: 10.1111/gtc.12183[Crossref], [PubMed], [CAS], Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXislSgsA%253D%253D&md5=061a2b69ebbb5d5f10a0ad17ce935c07Experimental design for stable genetic manipulation in mammalian cell lines: lentivirus and alternativesShearer, Robert F.; Saunders, Darren N.Genes to Cells (2015), 20 (1), 1-10CODEN: GECEFL; ISSN:1356-9597. (Wiley-Blackwell)A review. The use of third-generation lentiviral vectors is now commonplace in most areas of basic biol. These systems provide a fast, efficient means for modulating gene expression, but exptl. design needs to be carefully considered to minimize potential artifacts arising from off-target effects and other confounding factors. This review offers a starting point for those new to lentiviral-based vector systems, addressing the main issues involved with the use of lentiviral systems in vitro and outlines considerations which should be taken into account during exptl. design. Factors such as selecting an appropriate system and controls, and practical titrn. of viral transduction are important considerations for exptl. design. We also briefly describe some of the more recent advances in genome editing technol. TALENs and CRISPRs offer an alternative to lentivirus, providing endogenous gene editing with reduced off-target effects often at the expense of efficiency.
- 9Kohn, D. B.; Sadelain, M.; Glorioso, J. C. Occurrence of Leukaemia Following Gene Therapy of X-Linked SCID. Nat. Rev. Cancer. 2003, 3, 477– 488, DOI: 10.1038/nrc1122[Crossref], [PubMed], [CAS], Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXltVSqtrw%253D&md5=a51e02e95ee60bd85e3a30a4767d8b98Occurrence of leukemia following gene therapy of X-linked SCIDKohn, Donald B.; Sadelain, Michel; Glorioso, Joseph C.Nature Reviews Cancer (2003), 3 (7), 477-488CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)A review. Recombinant viral vectors have allowed gene transfer to be developed as a promising approach to the treatment of genetic diseases. Recently, gene therapy of children with X-linked severe combined immune deficiency resulted in impressive levels of immune reconstitution - a triumph that was later overshadowed by the development of leukemia in 2 patients. What were the causes of this cancer, and how can the therapeutic benefits of gene therapy be achieved while minimizing risk to the patient.
- 10Prescher, J. A.; Dube, D. H.; Bertozzi, C. R. Chemical Remodelling of Cell Surfaces in Living Animals. Nature. 2004, 430, 873– 877, DOI: 10.1038/nature02791[Crossref], [PubMed], [CAS], Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXmslCnu70%253D&md5=add5fdac7607d716a5564137ca6c06baChemical remodelling of cell surfaces in living animalsPrescher, Jennifer A.; Dube, Danielle H.; Bertozzi, Carolyn R.Nature (London, United Kingdom) (2004), 430 (7002), 873-877CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Cell surfaces are endowed with biol. functionality designed to mediate extracellular communication. The cell-surface repertoire can be expanded to include abiotic functionality through the biosynthetic introduction of unnatural sugars into cellular glycans, a process termed metabolic oligosaccharide engineering. This technique has been exploited in fundamental studies of glycan-dependent cell-cell and virus-cell interactions and also provides an avenue for the chem. remodelling of living cells. Unique chem. functional groups can be delivered to cell-surface glycans by metab. of the corresponding unnatural precursor sugars. These functional groups can then undergo covalent reaction with exogenous agents bearing complementary functionality. The exquisite chem. selectivity required of this process is supplied by the Staudinger ligation of azides and phosphines, a reaction that has been performed on cultured cells without detriment to their physiol. Here we demonstrate that the Staudinger ligation can be executed in living animals, enabling the chem. modification of cells within their native environment. The ability to tag cell-surface glycans in vivo may enable therapeutic targeting and non-invasive imaging of changes in glycosylation during disease progression.
- 11Baskin, J. M.; Prescher, J. A.; Laughlin, S. T.; Agard, N. J.; Chang, P. V.; Miller, I. A.; Lo, A.; Codelli, J. A.; Bertozzi, C. R. Copper-Free Click Chemistry for Dynamic in Vivo Imaging. Proc. National Acad. Sci. 2007, 104, 16793– 16797, DOI: 10.1073/pnas.0707090104[Crossref], [PubMed], [CAS], Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXht1KgtLvN&md5=7ea429c09b86509ff7f3960a95f11276Copper-free click chemistry for dynamic in vivo imagingBaskin, Jeremy M.; Prescher, Jennifer A.; Laughlin, Scott T.; Agard, Nicholas J.; Chang, Pamela V.; Miller, Isaac A.; Lo, Anderson; Codelli, Julian A.; Bertozzi, Carolyn R.Proceedings of the National Academy of Sciences of the United States of America (2007), 104 (43), 16793-16797CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Dynamic imaging of proteins in live cells is routinely performed by using genetically encoded reporters, an approach that cannot be extended to other classes of biomols. such as glycans and lipids. Here, the authors report a Cu-free variant of click chem. that can label these biomols. rapidly and selectively in living systems, overcoming the intrinsic toxicity of the canonical Cu-catalyzed reaction. The crit. reagent, a substituted cyclooctyne, possesses ring strain and electron-withdrawing fluorine substituents that together promote the [3+2] dipolar cycloaddn. with azides installed metabolically into biomols. This Cu-free click reaction possesses comparable kinetics to the Cu-catalyzed reaction and proceeds within minutes on live cells with no apparent toxicity. With this technique, the authors studied the dynamics of glycan trafficking and identified a population of sialoglycoconjugates with unexpectedly rapid internalization kinetics.
- 12Saxon, E.; Bertozzi, C. R. Cell Surface Engineering by a Modified Staudinger Reaction. Science. 2000, 287, 2007– 2010, DOI: 10.1126/science.287.5460.2007[Crossref], [PubMed], [CAS], Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXhvF2ru7g%253D&md5=91685aaba684aa4dfee86f44fef09886Cell surface engineering by a modified Staudinger reactionSaxon, Eliana; Bertozzi, Carolyn R.Science (Washington, D. C.) (2000), 287 (5460), 2007-2010CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Selective chem. reactions enacted within a cellular environment can be powerful tools for elucidating biol. processes or engineering novel interactions. A chem. transformation that permits the selective formation of covalent adducts among richly functionalized biopolymers within a cellular context is presented. A ligation modeled after the Staudinger reaction forms an amide bond by coupling of an azide and a specifically engineered triarylphosphine. Both reactive partners are abiotic and chem. orthogonal to native cellular components. Azides installed within cell surface glycoconjugates by metab. of a synthetic azidosugar were reacted with a biotinylated triarylphosphine to produce stable cell-surface adducts. The tremendous selectivity of the transformation should permit its execution within a cell's interior, offering new possibilities for probing intracellular interactions.
- 13Wang, X.; Lang, S.; Tian, Y.; Zhang, J.; Yan, X.; Fang, Z.; Weng, J.; Lu, N.; Wu, X.; Li, T.; Cao, H.; Li, Z.; Huang, X. Glycoengineering of Natural Killer Cells with CD22 Ligands for Enhanced Anticancer Immunotherapy. ACS Central Sci. 2020, 6, 382– 389, DOI: 10.1021/acscentsci.9b00956[ACS Full Text
], [CAS], Google Scholar
13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkt1yks70%253D&md5=5f6f36cf07a6f0428feb33487bfe6807Glycoengineering of Natural Killer Cells with CD22 Ligands for Enhanced Anticancer ImmunotherapyWang, Xianwu; Lang, Shuyao; Tian, Yunpeng; Zhang, Jianghong; Yan, Xu; Fang, Zhihong; Weng, Jian; Lu, Na; Wu, Xuanjun; Li, Tianlu; Cao, Hongzhi; Li, Zhu; Huang, XuefeiACS Central Science (2020), 6 (3), 382-389CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)Adoptive transfer of immune cells is being actively pursued for cancer treatment. Natural killer (NK) cells, a class of cytotoxic immune cells, generally lack inherent selectivities toward cancer. To bestow tumor-targeting abilities and enhance anticancer efficacy, a new strategy is established to glycoengineer NK cells. Carbohydrate-based ligands for CD22, a marker for B cell lymphoma, are introduced onto NK cells through either metabolic engineering or glyco-polymer insertion. Such NK cells exhibited greatly enhanced cytotoxicities toward CD22+ lymphoma cells in a CD22-dependent manner. Importantly, both CD22+ lymphoma cell lines and primary lymphoma cells from human cancer patients can be effectively killed by the engineered NK cells. Furthermore, glycoengineered NK cells provided significant protection to tumor-bearing mice. Thus, NK cell glycoengineering is an exciting new approach for cancer treatment complementing the current immune cell genetic engineering strategy. Sweetening natural killer cells enhanced their tumor-killing abilities in vitro and reduced tumor growth in vivo. - 14Frank, M. J.; Olsson, N.; Huang, A.; Tang, S.-W.; Negrin, R. S.; Elias, J. E.; Meyer, E. H. A Novel Antibody-Cell Conjugation Method to Enhance and Characterize Cytokine-Induced Killer Cells. Cytotherapy. 2020, 22, 135– 143, DOI: 10.1016/j.jcyt.2020.01.003[Crossref], [PubMed], [CAS], Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXltFykt7w%253D&md5=c61c8f677e77dd3c8edd1a4c3e24f9d4A novel antibody-cell conjugation method to enhance and characterize cytokine-induced killer cellsFrank, Matthew J.; Olsson, Niclas; Huang, Andy; Tang, Sai-Wen; Negrin, Robert S.; Elias, Joshua E.; Meyer, Everett H.Cytotherapy (2020), 22 (3), 135-143CODEN: CYTRF3; ISSN:1465-3249. (Elsevier Inc.)Cytokine-induced killer (CIK) cells are an ex vivo-expanded cellular therapy product with potent anti-tumor activity in a subset of patients with solid and hematol. malignancies. We hypothesize that directing CIK cells to a specific tumor antigen will enhance CIK cell anti-tumor cytotoxicity. We present a newly developed method for affixing antibodies directly to cell surface proteins. First, we evaluated the anti-tumor potential of CIK cells after affixing tumor-antigen targeting monoclonal antibodies. Second, we evaluated whether this antibody-conjugation method can profile the surface proteome of CIK cells. We demonstrated that affixing rituximab or daratumumab to CIK cells enhances cytotoxic killing of multiple lymphoma cell lines in vitro. These armed CIK cells exhibited enhanced intracellular signaling after engaging tumor targets. Cell surface proteome profiling suggested mechanisms by which antibody-armed CIK cells concurrently activated multiple surface proteins, leading to enhanced cytolytic activity. Our surface proteome anal. indicated that CIK cells display enhanced protein signatures indicative of immune responses, cellular activation and leukocyte migration. Here, we characterize the cell surface proteome of CIK cells using a novel methodol. that can be rapidly applied to other cell types. Without genetic modification CIK cells can be rapidly armed with monoclonal antibodies, which endows them with high specificity to kill tumor targets.
- 15Li, H.-K.; Hsiao, C.-W.; Yang, S.-H.; Yang, H.-P.; Wu, T.-S.; Lee, C.-Y.; Lin, Y.-L.; Pan, J.; Cheng, Z.-F.; Lai, Y.-D.; Hsiao, S.-C.; Tang, S.-W. A Novel Off-the-Shelf Trastuzumab-Armed NK Cell Therapy (ACE1702) Using Antibody-Cell-Conjugation Technology. Cancers. 2021, 13 (11), 2724, DOI: 10.3390/cancers13112724[Crossref], [PubMed], [CAS], Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXislehs7zO&md5=3945f97c94b8dbc432efd61eb8dfd07eA Novel off-the-Shelf Trastuzumab-Armed NK Cell Therapy (ACE1702) Using Antibody-Cell-Conjugation TechnologyLi, Hao-Kang; Hsiao, Ching-Wen; Yang, Sen-Han; Yang, Hsiu-Ping; Wu, Tai-Sheng; Lee, Chia-Yun; Lin, Yan-Liang; Pan, Janet; Cheng, Zih-Fei; Lai, Yan-Da; Hsiao, Shih-Chia; Tang, Sai-WenCancers (2021), 13 (11), 2724CODEN: CANCCT; ISSN:2072-6694. (MDPI AG)Simple Summary: Chimeric antigen receptor T cell therapy has shown its potency against hematol. malignancies in autologous settings but also limited success against solid tumors with severe adverse events, including fatal cases of cytokine releasing syndrome. The aim of this research is to develop a novel off-the-shelf natural killer cell therapy against HER2-expressing cancers using Antibody-Cell Conjugation (ACC) technol. and the endogenous CD16-expressing oNK cell line. ACE1702, trastuzumab-armed oNK cells with γ irradn. and cryopreservation, present superior in vitro and in vivo potency against HER2-expressing cancer cells and shows no tumorigenic potential, indicating the clin. application fighting HER2-expressing solid tumors. These findings suggest that ACC technol. can be applied to allogeneic immune cells to provide off-the-shelf therapies for cancer patients. Natural killer (NK) cells harbor efficient cytotoxicity against tumor cells without causing life-threatening cytokine release syndrome (CRS) or graft-vs.-host disease (GvHD). When compared to chimeric antigen receptor (CAR) technol., Antibody-Cell Conjugation (ACC) technol. has been developed to provide an efficient platform to arm immune cells with cancer-targeting antibodies to recognize and attack cancer cells. Recently, we established an endogenous CD16-expressing oNK cell line (oNK) with a favorable expression pattern of NK activation/inhibitory receptors. In this study, we applied ACC platform to conjugate oNK with trastuzumab and an anti-human epidermal growth factor receptor 2 (HER2) antibody. Trastuzumab-conjugated oNK, ACE-oNK-HER2, executed in vitro and in vivo cytotoxicity against HER2-expressing cancer cells and secretion of IFNγ. The irradiated and cryopreserved ACE-oNK-HER2, designated as ACE1702, retained superior HER2-specific in vitro and in vivo potency with no tumorigenic potential. In conclusion, this study provides the evidence to support the potential clin. application of ACE1702 as a novel off-the-shelf NK cell therapy against HER2-expressing solid tumors.
- 16Pishesha, N.; Bilate, A. M.; Wibowo, M. C.; Huang, N.-J.; Li, Z.; Deshycka, R.; Bousbaine, D.; Li, H.; Patterson, H. C.; Dougan, S. K.; Maruyama, T.; Lodish, H. F.; Ploegh, H. L. Engineered Erythrocytes Covalently Linked to Antigenic Peptides Can Protect against Autoimmune Disease. Proc. National Acad. Sci. 2017, 114, 3157– 3162, DOI: 10.1073/pnas.1701746114[Crossref], [PubMed], [CAS], Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjvVyjs7w%253D&md5=6a129adca06524e386953054bc3dd62eEngineered erythrocytes covalently linked to antigenic peptides can protect against autoimmune diseasePishesha, Novalia; Bilate, Angelina M.; Wibowo, Marsha C.; Huang, Nai-Jia; Li, Zeyang; Dhesycka, Rhogerry; Bousbaine, Djenet; Li, Hojun; Patterson, Heide C.; Dougan, Stephanie K.; Maruyama, Takeshi; Lodish, Harvey F.; Ploegh, Hidde L.Proceedings of the National Academy of Sciences of the United States of America (2017), 114 (12), 3157-3162CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Current therapies for autoimmune diseases rely on traditional immunosuppressive medications that expose patients to an increased risk of opportunistic infections and other complications. Immunoregulatory interventions that act prophylactically or therapeutically to induce antigen-specific tolerance might overcome these obstacles. Here the authors use the transpeptidase sortase to covalently attach disease-assocd. autoantigens to genetically engineered and to unmodified red blood cells as a means of inducing antigen-specific tolerance. This approach blunts the contribution to immunity of major subsets of immune effector cells (B cells, CD4+ and CD8+ T cells) in an antigen-specific manner. Transfusion of red blood cells expressing self-antigen epitopes can alleviate and even prevent signs of disease in exptl. autoimmune encephalomyelitis, as well as maintain normoglycemia in a mouse model of type 1 diabetes.
- 17Shi, J.; Kundrat, L.; Pishesha, N.; Bilate, A.; Theile, C.; Maruyama, T.; Dougan, S. K.; Ploegh, H. L.; Lodish, H. F. Engineered Red Blood Cells as Carriers for Systemic Delivery of a Wide Array of Functional Probes. Proc. National Acad. Sci. 2014, 111, 10131– 10136, DOI: 10.1073/pnas.1409861111[Crossref], [PubMed], [CAS], Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtVOit7vL&md5=0c8fc0c8a427682d0435231268bd71ddEngineered red blood cells as carriers for systemic delivery of a wide array of functional probesShi, Jiahai; Kundrat, Lenka; Pishesha, Novalia; Bilate, Angelina; Theile, Chris; Maruyama, Takeshi; Dougan, Stephanie K.; Ploegh, Hidde L.; Lodish, Harvey F.Proceedings of the National Academy of Sciences of the United States of America (2014), 111 (28), 10131-10136CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)We developed modified RBCs to serve as carriers for systemic delivery of a wide array of payloads. These RBCs contain modified proteins on their plasma membrane, which can be labeled in a sortase-catalyzed reaction under native conditions without inflicting damage to the target membrane or cell. Sortase accommodates a wide range of natural and synthetic payloads that allow modification of RBCs with substituents that cannot be encoded genetically. As proof of principle, we demonstrate site-specific conjugation of biotin to in vitro-differentiated mouse erythroblasts as well as to mature mouse RBCs. Thus modified, RBCs remain in the bloodstream for up to 28 d. A single domain antibody attached enzymically to RBCs enables them to bind specifically to target cells that express the antibody target. We extend these expts. to human RBCs and demonstrate efficient sortase-mediated labeling of in vitro-differentiated human reticulocytes.
- 18Harmand, T. J.; Pishesha, N.; Rehm, F. B. H.; Ma, W.; Pinney, W. B.; Xie, Y. J.; Ploegh, H. L. Asparaginyl Ligase-Catalyzed One-Step Cell Surface Modification of Red Blood Cells. ACS Chem. Biol. 2021, 16, 1201– 1207, DOI: 10.1021/acschembio.1c00216[ACS Full Text
], [CAS], Google Scholar
18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtlSmu7jI&md5=cdf20ff3ecfc82c4b1b50bb019c90306Asparaginyl Ligase-Catalyzed One-Step Cell Surface Modification of Red Blood CellsHarmand, Thibault J.; Pishesha, Novalia; Rehm, Fabian B. H.; Ma, Weiyi; Pinney, William B.; Xie, Yushu J.; Ploegh, Hidde L.ACS Chemical Biology (2021), 16 (7), 1201-1207CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Red blood cells (RBCs) can serve as vascular carriers for drugs, proteins, peptides, and nanoparticles. Human RBCs remain in the circulation for ∼120 days, are biocompatible, and are immunol. largely inert. RBCs are cleared by the reticuloendothelial system and can induce immune tolerance to foreign components attached to the RBC surface. RBC conjugates have been pursued in clin. trials to treat cancers and autoimmune diseases and to correct genetic disorders. Still, most methods used to modify RBCs require multiple steps, are resource-intensive and time-consuming, and increase the risk of inflicting damage to the RBCs. Here, we describe direct conjugation of peptides and proteins onto the surface of RBCs in a single step, catalyzed by a highly efficient, recombinant asparaginyl ligase under mild, physiol. conditions. In mice, the modified RBCs remain intact in the circulation, display a normal circulatory half-life, and retain their immune tolerance-inducing properties, as shown for protection against an accelerated model for type 1 diabetes. We conjugated different nanobodies to RBCs with retention of their binding properties, and these modified RBCs can target cancer cells in vitro. This approach provides an appealing alternative to current methods of RBC engineering. It provides ready access to more complex RBC constructs and highlights the general utility of asparaginyl ligases for the modification of native cell surfaces. - 19Li, J.; Chen, M.; Liu, Z.; Zhang, L.; Felding, B. H.; Moremen, K. W.; Lauvau, G.; Abadier, M.; Ley, K.; Wu, P. A Single-Step Chemoenzymatic Reaction for the Construction of Antibody–Cell Conjugates. ACS Central Sci. 2018, 4, 1633– 1641, DOI: 10.1021/acscentsci.8b00552[ACS Full Text
], [CAS], Google Scholar
19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisVaqsrbP&md5=59cb733e3db9f5b9ffb711451afd68b4A Single-Step Chemoenzymatic Reaction for the Construction of Antibody-Cell ConjugatesLi, Jie; Chen, Mingkuan; Liu, Zilei; Zhang, Linda; Felding, Brunie H.; Moremen, Kelley W.; Lauvau, Gregoire; Abadier, Michael; Ley, Klaus; Wu, PengACS Central Science (2018), 4 (12), 1633-1641CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)Employing live cells as therapeutics is a direction of future drug discovery. An easy and robust method to modify the surfaces of cells directly to incorporate novel functionalities is highly desirable. However, genetic methods for cell-surface engineering are laborious and limited by low efficiency for primary cell modification. Here we report a chemoenzymic approach that exploits a fucosyltransferase to transfer bio-macromols., such as an IgG antibody (MW∼ 150 kDa), to the glycocalyx on the surfaces of live cells when the antibody is conjugated to the enzyme's natural donor substrate GDP-Fucose. Requiring no genetic modification, this method is fast and biocompatible with little interference to cells' endogenous functions. We applied this method to construct two antibody-cell conjugates (ACCs) using both cell lines and primary cells, and the modified cells exhibited specific tumor targeting and resistance to inhibitory signals produced by tumor cells, resp. Remarkably, Herceptin-NK-92MI conjugates, a natural killer cell line modified with Herceptin, exhibit enhanced activities to induce the lysis of HER2+ cancer cells both ex vivo and in a human tumor xenograft model. Given the unprecedented substrate tolerance of the fucosyltransferase, this chemoenzymic method offers a general approach to engineer cells as research tools and for therapeutic applications. - 20Maza, J. C.; Bader, D. L. V.; Xiao, L.; Marmelstein, A. M.; Brauer, D. D.; ElSohly, A. M.; Smith, M. J.; Krska, S. W.; Parish, C. A.; Francis, M. B. Enzymatic Modification of N-Terminal Proline Residues Using Phenol Derivatives. J. Am. Chem. Soc. 2019, 141, 3885– 3892, DOI: 10.1021/jacs.8b10845[ACS Full Text
], [CAS], Google Scholar
20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXisFyitbc%253D&md5=8b203591be06bb432717258ea0452ca3Enzymatic Modification of N-Terminal Proline Residues Using Phenol DerivativesMaza, Johnathan C.; Bader, Daniel L. V.; Xiao, Lifeng; Marmelstein, Alan M.; Brauer, Daniel D.; El Sohly, Adel M.; Smith, Matthew J.; Krska, Shane W.; Parish, Craig A.; Francis, Matthew B.Journal of the American Chemical Society (2019), 141 (9), 3885-3892CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)A convenient enzymic strategy is reported for the modification of proline residues in the N-terminal positions of proteins. Using a tyrosinase enzyme isolated from Agaricus bisporus (abTYR), phenols and catechols are oxidized to highly reactive o-quinone intermediates that then couple to N-terminal proline residues in high yield. Key advantages of this bioconjugation method include (1) the use of air-stable precursors that can be prepd. on large scale if needed, (2) mild reaction conditions, including low temps., (3) the targeting of native functional groups that can be introduced readily on most proteins, and (4) the use of mol. oxygen as the sole oxidant. This coupling strategy was successfully demonstrated for the attachment of a variety of phenol-derivatized cargo mols. to a series of protein substrates, including self-assembled viral capsids, enzymes, and a chitin binding domain (CBD). The ability of the CBD to bind to the surfaces of yeast cells was found to be unperturbed by this modification reaction. - 21Lobba, M. J.; Fellmann, C.; Marmelstein, A. M.; Maza, J. C.; Kissman, E. N.; Robinson, S. A.; Staahl, B. T.; Urnes, C.; Lew, R. J.; Mogilevsky, C. S.; Doudna, J. A.; Francis, M. B. Site-Specific Bioconjugation through Enzyme-Catalyzed Tyrosine–Cysteine Bond Formation. ACS Central Sci. 2020, 6, 1564– 1571, DOI: 10.1021/acscentsci.0c00940[ACS Full Text
], [CAS], Google Scholar
21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs1eltL3P&md5=a0fe6337ef9daa4caa637a7ba54f9d68Site-Specific Bioconjugation through Enzyme-Catalyzed Tyrosine-Cysteine Bond FormationLobba, Marco J.; Fellmann, Christof; Marmelstein, Alan M.; Maza, Johnathan C.; Kissman, Elijah N.; Robinson, Stephanie A.; Staahl, Brett T.; Urnes, Cole; Lew, Rachel J.; Mogilevsky, Casey S.; Doudna, Jennifer A.; Francis, Matthew B.ACS Central Science (2020), 6 (9), 1564-1571CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)The synthesis of protein-protein and protein-peptide conjugates is an important capability for producing vaccines, immunotherapeutics, and targeted delivery agents. Herein we show that the enzyme tyrosinase is capable of oxidizing exposed tyrosine residues into o-quinones that react rapidly with cysteine residues on target proteins. This coupling reaction occurs under mild aerobic conditions and has the rare ability to join full-size proteins in under 2 h. The utility of the approach is demonstrated for the attachment of cationic peptides to enhance the cellular delivery of CRISPR-Cas9 20-fold and for the coupling of reporter proteins to a cancer-targeting antibody fragment without loss of its cell-specific binding ability. The broad applicability of this technique provides a new building block approach for the synthesis of protein chimeras. Enzymic oxidn. of tyrosine residues followed by reaction with cysteine thiols allows the covalent coupling of proteins and peptides using only native amino acid side chains. - 22Marmelstein, A. M.; Lobba, M. J.; Mogilevsky, C. S.; Maza, J. C.; Brauer, D. D.; Francis, M. B. Tyrosinase-Mediated Oxidative Coupling of Tyrosine Tags on Peptides and Proteins. J. Am. Chem. Soc. 2020, 142, 5078– 5086, DOI: 10.1021/jacs.9b12002[ACS Full Text
], [CAS], Google Scholar
22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXjs12gtLc%253D&md5=e61c218de8ccf478384e3fbbb3407f65Tyrosinase-Mediated Oxidative Coupling of Tyrosine Tags on Peptides and ProteinsMarmelstein, Alan M.; Lobba, Marco J.; Mogilevsky, Casey S.; Maza, Johnathan C.; Brauer, Daniel D.; Francis, Matthew B.Journal of the American Chemical Society (2020), 142 (11), 5078-5086CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Oxidative coupling (OC) through o-quinone intermediates has been established as an efficient and site-selective way to modify protein N-termini and the unnatural amino acid p-aminophenylalanine (paF). Recently, we reported that the tyrosinase-mediated oxidn. of phenol-tagged cargo mols. is a particularly convenient method of generating o-quinones in situ. The coupling partners can be easily prepd. and stored, the reaction takes place under mild conditions (phosphate buffer, pH 6.5, 4 to 23 °C), and dissolved oxygen is the only oxidant required. Here, we show an important extension of this chem. for the activation of tyrosine residues that project into soln. from the N or C-termini of peptide and protein substrates. Generating the o-quinone electrophiles from tyrosine allows greater flexibility in choosing the nucleophilic coupling partner and expands the scope of the reaction to include C-terminal positions. We also introduce a new bacterial tyrosinase enzyme that shows improved activation for some tyrosine substrates. The efficacy of several secondary amines and aniline derivs. was evaluated in the coupling reactions, providing important information for coupling partner design. This strategy was used to modify the C-termini of an antibody scFv construct and of Protein L, a human IgG kappa light chain binding protein. The use of the modified proteins as immunolabeling agents was also demonstrated. - 23Bruins, J. J.; Westphal, A. H.; Albada, B.; Wagner, K.; Bartels, L.; Spits, H.; van Berkel, W. J. H.; van Delft, F. L. Inducible, Site-Specific Protein Labeling by Tyrosine Oxidation–Strain-Promoted (4 + 2) Cycloaddition. Bioconjugate Chem. 2017, 28, 1189– 1193, DOI: 10.1021/acs.bioconjchem.7b00046[ACS Full Text
], [CAS], Google Scholar
23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjs12ntro%253D&md5=058655ebcbdef65719b9a0613dae3d51Inducible, Site-Specific Protein Labeling by Tyrosine Oxidation-Strain-Promoted (4 + 2) CycloadditionBruins, Jorick J.; Westphal, Adrie H.; Albada, Bauke; Wagner, Koen; Bartels, Lina; Spits, Hergen; van Berkel, Willem J. H.; van Delft, Floris L.Bioconjugate Chemistry (2017), 28 (4), 1189-1193CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Genetically encoded tyrosine (Y-tag) can be utilized as a latent anchor for inducible and site-selective conjugation. Upon oxidn. of tyrosine with mushroom tyrosinase, strain-promoted cycloaddn. (SPOCQ) of the resulting 1,2-quinone with various bicyclo[6.1.0]nonyne (BCN) derivs. led to efficient conjugation. The method was applied for fluorophore labeling of laminarinase A and for the site-specific prepn. of an antibody-drug conjugate. - 24Wilton, E. E.; Opyr, M. P.; Kailasam, S.; Kothe, R. F.; Wieden, H.-J. SdAb-DB: The Single Domain Antibody Database. ACS Synth. Biol. 2018, 7, 2480– 2484, DOI: 10.1021/acssynbio.8b00407[ACS Full Text
], [CAS], Google Scholar
24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitFertbbJ&md5=991b0b08079a4f778d8150b902fb010csdAb-DB: The Single Domain Antibody DatabaseWilton, Emily E.; Opyr, Michael P.; Kailasam, Senthilkumar; Kothe, Ronja F.; Wieden, Hans-JoachimACS Synthetic Biology (2018), 7 (11), 2480-2484CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)A review. The Single Domain Antibody Database, or sdAb-DB, (www.sdab-db.ca) is the first freely available repository for single domain antibodies and related classes of proteins. Due to their small size, modular structure, and ease of expression, single domain antibodies (sdAb) have a wide range of applications, including as a rational design tool, and are therefore of great interest for synthetic biologists and bioengineers. However, to enable effective use and sharing of existing sdAbs, including those with engineered functions (e.g., fusions with fluorescent proteins), as well as the rational design and engineering of new sdAbs, it is necessary to have access to sequences and exptl. data. We have therefore developed a publicly available, sdAb-focused database, providing access to manually curated sdAb data from protein databases, published scientific articles, and user submissions. The sdAb-DB is an open-source repository and sharing platform for the sdAb community, providing access to performance data and basic bioinformatic tools for use with previously described and validated sdAbs, as well as for the engineering of new sdAb-based designs and proteins. - 25Kirchhofer, A.; Helma, J.; Schmidthals, K.; Frauer, C.; Cui, S.; Karcher, A.; Pellis, M.; Muyldermans, S.; Casas-Delucchi, C. S.; Cardoso, M. C.; Leonhardt, H.; Hopfner, K.-P.; Rothbauer, U. Modulation of Protein Properties in Living Cells Using Nanobodies. Nat. Struct. Mol. Biol. 2010, 17, 133– 138, DOI: 10.1038/nsmb.1727[Crossref], [PubMed], [CAS], Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsFGjtr3K&md5=893913260f608a5337037fc43891ba24Modulation of protein properties in living cells using nanobodiesKirchhofer, Axel; Helma, Jonas; Schmidthals, Katrin; Frauer, Carina; Cui, Sheng; Karcher, Annette; Pellis, Mireille; Muyldermans, Serge; Casas-Delucchi, Corella S.; Cardoso, M. Cristina; Leonhardt, Heinrich; Hopfner, Karl-Peter; Rothbauer, UlrichNature Structural & Molecular Biology (2010), 17 (1), 133-138CODEN: NSMBCU; ISSN:1545-9993. (Nature Publishing Group)Protein conformation is critically linked to function and often controlled by interactions with regulatory factors. Here we report the selection of camelid-derived single-domain antibodies (nanobodies) that modulate the conformation and spectral properties of the green fluorescent protein (GFP). One nanobody could reversibly reduce GFP fluorescence by a factor of 5, whereas its displacement by a second nanobody caused an increase by a factor of 10. Structural anal. of GFP-nanobody complexes revealed that the two nanobodies induce subtle opposing changes in the chromophore environment, leading to altered absorption properties. Unlike conventional antibodies, the small, stable nanobodies are functional in living cells. Nanobody-induced changes were detected by ratio imaging and used to monitor protein expression and subcellular localization as well as translocation events such as the tamoxifen-induced nuclear localization of estrogen receptor (ER). This work demonstrates that protein conformations can be manipulated and studied with nanobodies in living cells.
- 26Morvan, M. G.; Lanier, L. L. NK Cells and Cancer: You Can Teach Innate Cells New Tricks. Nat. Rev. Cancer. 2016, 16, 7– 19, DOI: 10.1038/nrc.2015.5[Crossref], [PubMed], [CAS], Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXitVOjsbnE&md5=bc3ca6826c04df6147dbe9770b90f420NK cells and cancer: you can teach innate cells new tricksMorvan, Maelig G.; Lanier, Lewis L.Nature Reviews Cancer (2016), 16 (1), 7-19CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)Natural killer (NK) cells are the prototype innate lymphoid cells endowed with potent cytolytic function that provide host defense against microbial infection and tumors. Here, we review evidence for the role of NK cells in immune surveillance against cancer and highlight new therapeutic approaches for targeting NK cells in the treatment of cancer.
- 27Bald, T.; Krummel, M. F.; Smyth, M. J.; Barry, K. C. The NK Cell–Cancer Cycle: Advances and New Challenges in NK Cell–Based Immunotherapies. Nat. Immunol. 2020, 21, 835– 847, DOI: 10.1038/s41590-020-0728-z[Crossref], [PubMed], [CAS], Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVWgt7fK&md5=b47b77228617a9253f7b5dd80319bd23The NK cell-cancer cycle: advances and new challenges in NK cell-based immunotherapiesBald, Tobias; Krummel, Matthew F.; Smyth, Mark J.; Barry, Kevin C.Nature Immunology (2020), 21 (8), 835-847CODEN: NIAMCZ; ISSN:1529-2908. (Nature Research)A review. Abstr.: Natural killer (NK) cells belong to the innate immune system and contribute to protecting the host through killing of infected, foreign, stressed or transformed cells. Addnl., via cellular cross-talk, NK cells orchestrate antitumor immune responses. Hence, significant efforts have been undertaken to exploit the therapeutic properties of NK cells in cancer. Current strategies in preclin. and clin. development include adoptive transfer therapies, direct stimulation, recruitment of NK cells into the tumor microenvironment (TME), blockade of inhibitory receptors that limit NK cell functions, and therapeutic modulation of the TME to enhance antitumor NK cell function. In this Review, we introduce the NK cell-cancer cycle to highlight recent advances in NK cell biol. and to discuss the progress and problems of NK cell-based cancer immunotherapies.
- 28Tam, Y. K.; Maki, G.; Miyagawa, B.; Hennemann, B.; Tonn, T.; Klingemann, H.-G. Characterization of Genetically Altered, Interleukin 2-Independent Natural Killer Cell Lines Suitable for Adoptive Cellular Immunotherapy. Hum. Gene Ther. 1999, 10, 1359– 1373, DOI: 10.1089/10430349950018030[Crossref], [PubMed], [CAS], Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXjsVyqtbs%253D&md5=1c51fc49a1b3c58426dc16e1953f3785Characterization of genetically altered, interleukin 2-independent natural killer cell lines suitable for adoptive cellular immunotherapyTam, Y. K.; Maki, G.; Miyagawa, B.; Hennemann, B.; Tonn, T.; Klingemann, H.-G.Human Gene Therapy (1999), 10 (8), 1359-1373CODEN: HGTHE3; ISSN:1043-0342. (Mary Ann Liebert, Inc.)NK-92 is a highly cytotoxic natural killer (NK) tumor cell line that possesses properties that make it an excellent candidate for adoptive cellular immunotherapy. However, the cytotoxicity of NK cells is dependent on cytokines such as interleukin 2 (IL-2). Although NK-92 cells maintain cytotoxicity for a time after withdrawal of IL-2, clin. use will probably require prolonged treatment with fully activated cells to eliminate disease effectively. The ability to support cytotoxic cells with exogenously administered IL-2 is limited by assocd. toxicity. Therefore, the authors describe the transfection of the IL-2-dependent NK-92 cell line with human IL-2 (hIL-2) cDNA by particle-mediated gene transfer to create two IL-2-independent variants, NK-92MI and NK-92CI, and describe their characterization and comparison with parental cells. Both variants were shown to contain, express, and synthesize the hIL-2 cDNA. IL-2 synthesis was higher in NK-92MI cells compared with NK-92CI cells, with no expression in parental cells. Functionally, the cytotoxicity of all three cell lines was similar and coincubation with IL-2-independent variants did not affect hematopoietic progenitor cells. NK-92MI and NK-92CI cells were more radiosensitive than NK-92 cells, with proliferation inhibited at lower radiation doses and increased morality and decreased cytotoxicity compared with parental cells. Data presented here show that the authors have created by particle-mediated gene transfer two IL-2-independent variants of NK-92 that are identical to parental cells in virtually all respects, including high cytotoxic activity. The nonviral transfection of these cells makes them suitable for clin. applications. These IL-2-independent cells should allow prolonged treatment with fully active natural killer cells without the need for exogenous IL-2 support.
- 29Suck, G.; Odendahl, M.; Nowakowska, P.; Seidl, C.; Wels, W. S.; Klingemann, H. G.; Tonn, T. NK-92: An ‘off-the-Shelf Therapeutic’ for Adoptive Natural Killer Cell-Based Cancer Immunotherapy. Cancer Immunol. Immunother. 2016, 65, 485– 492, DOI: 10.1007/s00262-015-1761-x[Crossref], [PubMed], [CAS], Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvVeqsb7L&md5=2ba14aed3365a23ad62211242c03a2aaNK-92: an 'off-the-shelf therapeutic' for adoptive natural killer cell-based cancer immunotherapySuck, Garnet; Odendahl, Marcus; Nowakowska, Paulina; Seidl, Christian; Wels, Winfried S.; Klingemann, Hans G.; Tonn, TorstenCancer Immunology Immunotherapy (2016), 65 (4), 485-492CODEN: CIIMDN; ISSN:0340-7004. (Springer)Natural killer (NK) cells are increasingly considered as immunotherapeutic agents in particular in the fight against cancers. NK cell therapies are potentially broadly applicable and, different from their T cell counterparts, do not cause graft-vs.-host disease. Efficacy and clin. in vitro or in vivo expansion of primary NK cells will however always remain variable due to individual differences of donors or patients. Long-term storage of clin. NK cell lots to allow repeated clin. applications remains an addnl. challenge. In contrast, the established and well-characterized cell line NK-92 can be easily and reproducibly expanded from a good manufg. practice (GMP)-compliant cryopreserved master cell bank. Moreover, no cost-intensive cell purifn. methods are required. To date, NK-92 has been intensively studied. The cells displayed superior cytotoxicity against a no. of tumor types tested, which was confirmed in preclin. mouse studies. Subsequent clin. testing demonstrated safety of NK-92 infusions even at high doses. Despite the phase I nature of the trials conducted so far, some efficacy was noted, particularly against lung tumors. Furthermore, to overcome tumor resistance and for specific targeting, NK-92 has been engineered to express a no. of different chimeric antigen receptors (CARs), including targeting, for example, CD19 or CD20 (anti-B cell malignancies), CD38 (anti-myeloma) or human epidermal growth factor receptor 2 (HER2; ErbB2; anti-epithelial cancers). The concept of an NK cell line as an allogeneic cell therapeutic produced 'off-the-shelf' on demand holds great promise for the development of effective treatments.
- 30Obermeyer, A. C.; Jarman, J. B.; Francis, M. B. N-Terminal Modification of Proteins with O-Aminophenols. J. Am. Chem. Soc. 2014, 136, 9572– 9579, DOI: 10.1021/ja500728c[ACS Full Text
], [CAS], Google Scholar
30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtVGlu77L&md5=ab16257753b27283d65e3d787b0bff7fN-Terminal Modification of Proteins with o-AminophenolsObermeyer, Allie C.; Jarman, John B.; Francis, Matthew B.Journal of the American Chemical Society (2014), 136 (27), 9572-9579CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The synthetic modification of proteins plays an important role in chem. biol. and biomaterials science. These fields provide a const. need for chem. tools that can introduce new functionality in specific locations on protein surfaces. In this work, an oxidative strategy is demonstrated for the efficient modification of N-terminal residues on peptides and N-terminal proline residues on proteins. The strategy uses o-aminophenols or o-catechols that are oxidized to active coupling species in situ using potassium ferricyanide. Peptide screening results have revealed that many N-terminal amino acids can participate in this reaction, and that proline residues are particularly reactive. When applied to protein substrates, the reaction shows a stronger requirement for the proline group. Key advantages of the reaction include its fast second-order kinetics and ability to achieve site-selective modification in a single step using low concns. of reagent. Although free cysteines are also modified by the coupling reaction, they can be protected through disulfide formation and then liberated after N-terminal coupling is complete. This allows access to doubly functionalized bioconjugates that can be difficult to access using other methods. - 31Hurben, A. K.; Erber, L. N.; Tretyakova, N. Y.; Doran, T. M. Proteome-Wide Profiling of Cellular Targets Modified by Dopamine Metabolites Using a Bio-Orthogonally Functionalized Catecholamine. ACS Chem. Biol. 2021, 16, 2581– 2594, DOI: 10.1021/acschembio.1c00629[ACS Full Text
], [CAS], Google Scholar
31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitlOhur%252FF&md5=a4b345179f27c8e8b385dbcaffbb3b64Proteome-Wide Profiling of Cellular Targets Modified by Dopamine Metabolites Using a Bio-Orthogonally Functionalized CatecholamineHurben, Alexander K.; Erber, Luke N.; Tretyakova, Natalia Y.; Doran, Todd M.ACS Chemical Biology (2021), 16 (11), 2581-2594CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Selective death of midbrain dopaminergic neurons is a hallmark pathol. of Parkinson's disease (PD), but the mol. mechanisms that initiate the cascade of events resulting in neurodegeneration in PD remain unclear. Compelling evidence suggests that dysregulation of dopamine (DA) induces neuronal stress and damage responses that are operative processes in striatal degeneration preceding PD-like symptoms. Improper DA sequestration to vesicles raises cytosolic DA levels, which is rapidly converted into electrophilic dopaquinone species (DQs) that react readily with protein nucleophiles forming covalent modifications that alter the native structure and function of proteins. These so-called DA-protein adducts (DPAs) have been reported to play a role in neurotoxicity, and their abundance with respect to neurodegeneration has been linked to clin. and pathol. features of PD that suggest that they play a causal role in PD pathogenesis. Therefore, characterizing DPAs is a crit. first step in understanding the susceptibility of midbrain dopaminergic neurons during PD. To help achieve this goal, we report here a novel DA-mimetic (DAyne) contg. a biorthogonal alkyne handle that exhibits a reactivity profile similar to DA in aq. buffers. By linking DPAs formed with DAyne to a fluorescent reporter mol., DPAs were visualized in fixed cells and within lysates. DAyne enabled global mapping of cellular proteins affected by DQ modification and their bioactive pathways through enrichment. Our proteomic profiling of DPAs in neuronal SH-SY5Y cells indicates that proteins susceptible to DPA formation are extant throughout the proteome, potentially influencing several diverse biol. pathways involved in PD such as endoplasmic reticulum (ER) stress, cytoskeletal instability, proteotoxicity, and clathrin function. We validated that a protein involved in the ER stress pathway, protein disulfide isomerase 3 (PDIA3), which was enriched in our chemoproteomic anal., is functionally inhibited by DA, providing evidence that dysregulated cellular DA may induce or exacerbate ER stress. Thus, DAyne provided new mechanistic insights into DA toxicity that may be obsd. during PD by enabling characterization of DPAs generated reproducibly at physiol. relevant quinone exposures. We anticipate our design and application of this reactivity-based probe will be generally applicable for clarifying mechanisms of metabolic quinone toxicity. - 32Bruce, V. J.; Lopez-Islas, M.; McNaughton, B. R. Resurfaced Cell-Penetrating Nanobodies: A Potentially General Scaffold for Intracellularly Targeted Protein Discovery. Protein Sci. 2016, 25, 1129– 1137, DOI: 10.1002/pro.2926[Crossref], [PubMed], [CAS], Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XltlWhsrc%253D&md5=849322d18d9dea14cd01aa3977f7b72bResurfaced cell-penetrating nanobodies: A potentially general scaffold for intracellularly targeted protein discoveryBruce, Virginia J.; Lopez-Islas, Monica; McNaughton, Brian R.Protein Science (2016), 25 (6), 1129-1137CODEN: PRCIEI; ISSN:1469-896X. (Wiley-Blackwell)By virtue of their size, functional group diversity, and complex structure, proteins can often recognize and modulate disease-relevant macromols. that present a challenge to small-mol. reagents. Addnl., high-throughput screening and evolution-based methods often make the discovery of new protein binders simpler than the analogous small-mol. discovery process. However, most proteins do not cross the lipid bilayer membrane of mammalian cells. This largely limits the scope of protein therapeutics and basic research tools to those targeting disease-relevant receptors on the cell surface or extracellular matrix. Previously, researchers have shown that cationic resurfacing of proteins can endow cell penetration. However, in our experience, many proteins are not amenable to such extensive mutagenesis. Here, we report that nanobodies-a small and stable protein that can be evolved to recognize virtually any disease-relevant receptor-are amenable to cationic resurfacing, which results in cell internalization. Once internalized, these nanobodies access the cytosol. Polycationic resurfacing does not appreciably alter the structure, expression, and function (target recognition) of a previously reported GFP-binding nanobody, and multiple nanobody scaffolds are amenable to polycationic resurfacing. Given this, we propose that polycationic resurfaced cell-penetrating nanobodies might represent a general scaffold for intracellularly targeted protein drug discovery.
- 33Lyon, R. Drawing Lessons from the Clinical Development of Antibody-Drug Conjugates. Drug Discovery Today Technologies. 2018, 30, 105– 109, DOI: 10.1016/j.ddtec.2018.10.001[Crossref], [PubMed], [CAS], Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cnisVSlsA%253D%253D&md5=dfdb2af91ce423b8912dc33f9187b35fDrawing lessons from the clinical development of antibody-drug conjugatesLyon RobertDrug discovery today. Technologies (2018), 30 (), 105-109 ISSN:.The antibody-drug conjugate (ADC) field has seen a remarkable expansion in the number of entrants in clinical studies. Many of these agents employ newer conjugation technologies that have been developed over the last decade that confer various attributes to the ADCs prepared with them, including stability, potency, and homogeneity. In many cases, these new ADCs appear demonstrably superior to earlier technologies in preclinical models of activity and toxicology, but the degree to which these improvements will translate to the clinic is only starting to be seen. Many of these technologies are now competing head-to-head by targeting the same antigen in similar patient populations, allowing for a direct comparison of their clinical performance properties. As lessons from these experiences feed back into discovery research, future iterations of ADC design may be expected to bring improved therapeutics into the clinic.
- 34Bauer, S.; Groh, V.; Wu, J.; Steinle, A.; Phillips, J. H.; Lanier, L. L.; Spies, T. Activation of NK Cells and T Cells by NKG2D, a Receptor for Stress-Inducible MICA. Science. 1999, 285, 727– 729, DOI: 10.1126/science.285.5428.727[Crossref], [PubMed], [CAS], Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXltVehsro%253D&md5=296b66b9314dcaaa0bb23d448df2e186Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICABauer, Stefan; Groh, Veronika; Phillips, Joseph H.; Lainer, Lewis L.; Spies, ThomasScience (Washington, D. C.) (1999), 285 (5428), 727-729CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Stress-inducible MICA, a distant homolog of major histocompatibility complex (MHC) class I, functions as an antigen for γδ T cells and is frequently expressed in epithelial tumors. A receptor for MICA was detected on most γδ T cells, CD8+ αβ T cells, and natural killer (NK) cells and was identified as NKG2D. Effector cells from all these subsets could be a stimulated by ligation of NKG2D. Engagement of NKG2D activated cytolytic responses of γδ T cells and NK cells against transfectants and epithelial tumor cells expressing MICA. These results define an activating immunoreceptor-MHC ligand interaction that may promote antitumor NK and T cell responses.
Cited By
This article is cited by 2 publications.
- Jing Dai, Kiera B. Wilhelm, Amanda J. Bischoff, Jose H. Pereira, Michel T. Dedeo, Derek M. García‐Almedina, Paul D. Adams, Jay T. Groves, Matthew B. Francis. A Membrane‐Associated Light‐Harvesting Model is Enabled by Functionalized Assemblies of Gene‐Doubled TMV Proteins. Small 2023, 3 , 2207805. https://doi.org/10.1002/smll.202207805
- Qiong Liu, Xinxin Ren, Yufang Hu, Jieying Zhou. Versatile Electrochemical Platform for GSH Detection and its Boolean Logic Application in Related Biological Pathways. Journal of The Electrochemical Society 2022, 169 (12) , 127516. https://doi.org/10.1149/1945-7111/aca8d7
Abstract
Figure 1
Figure 1. General strategy for modifying cell surfaces with nanobodies. (a) Tyrosinase catalyzes the oxidation of small-molecule phenols to highly reactive o-quinones, which can modify nucleophiles present on proteins. Engineered tyrosine tags at protein termini can also be oxidized by tyrosinase, producing a site-specific o-quinone on the protein that reacts with protein-based nucleophiles. (b) Tyrosine-tagged nanobodies can be site-specifically oxidized by tyrosinase for attachment of these proteins to cells. The resulting linkage produces a well-defined point of attachment for installing nanobodies on cell surfaces while imbuing the target cell with novel antigen-binding functionality. (c) Nanobodies are low-molecular-weight (∼10–15 kDa) antigen-binders derived from the variable region of the camelid antibody (PDB ID 3K1K).
Figure 2
Figure 2. Modification of NK cell surfaces with nanobodies. (a) Tyrosinase enzyme produces a site-specific o-quinone at the C-terminal Ser–Gly4–Tyr tag installed on nanobodies, as evidenced by a 14 Da mass shift detected via ESI-TOF MS. (b) To verify that NK cell surfaces can be decorated with nanobodies using tyorsinase, a Tyr-tagged nanobody against GFP (nbGFPTyr) was designed. Using tyrosinase, nbGFPTyr can be attached to the cell surface, and 2° labeling with GFP can be used to analyze the reaction using flow cytometry. (c) Labeling experiments with nbGFPTyr validated attachment of the nanobody to the cell surface, as only cells treated with both nbGFPTyr and tyrosinase showed an increase in GFP fluorescence (red trace) over controls (blue and orange traces). (d) Using a Cys point mutant, a single FITC dye can be attached to each nbGFPTyr (nbFITC). After attachment of 10 μM nbFITC to cell surfaces, comparison against FITC-calibration beads determined that a median value of ∼120,000 copies of the nanobody were linked to the cells. Data are represented as box plots, with the top of the box representing the 75th percentile of the data, the middle line representing the median of the data, and the bottom of the box representing the 25th percentile of the data.
Figure 3
Figure 3. Decoration of NK cells for nanobody-directed cell–cell interactions. (a) Using tyrosinase, a Tyr-tagged nanobody against HER2 (nbHER2Tyr) was attached to NK cells. Secondary labeling with a soluble FITC–HER2 showed that only cells exposed to nbHER2Tyr and tyrosinase exhibited a shift in FITC signal detected via flow cytometry (red trace) over controls. (b) To assess if tyrosinase-synthesized NK–nbHER2 conjugates can make targeted contacts with HER2+ cells, NK–nbHER2 cells were mixed with a HER2+ cell line (SK-BR-3) at a ratio of 2:1 (NK:target). Cells were allowed to bind and settle and then imaged using fluorscence microscopy. A nearest neighbor analysis was performed (CellProfiler), indicating that a statistically significant proportion of target cells (green) were bound to two or more NK–nbHER2 cells (red) only when the NK cells were pretreated with nbHER2Tyr and tyrosinase (orange bar). (c) Fluorescence microscopy images confirm rosette formation is only seen when NK cells are pretreated with both nbHER2Tyr and tyrosinase.
Figure 4
Figure 4. Targeted cell killing elicited by tyrosinase-synthesized nanobody–NK cell conjugates. (a) Schematic representation of the fluorescence-based cell assay used to determine NK cytotoxicity. HER2+ cells (SK-BR-3) were preloaded with calcein AM dye, which is retained by the cell membrane after uptake. Lysis of the HER2+ cell releases dye into the supernatant, providing a measurement for cell lysis. Only NK cells pretreated with both nbHER2Tyr and tyrosinase (orange bar) show statistically significant specific cell lysis over control treatments. (b) To assess how the ratio of NK:target cell impacts specific cytotoxicity, NK–nbHER2 cells were synthesized using 10 μM nbHER2Tyr and 400 nM tyrosinase and mixed with calcein AM loaded HER2+ cells (SK-BR-3). Statistically significant cell death was observed at ratios even as low as 2:1 (effector:target). (c) To assess the required concentration of nbHER2Tyr needed to elict NK-mediated cell death, a variety of concentrations of nbHER2Tyr were used to label NK cells with tyrosinase. Increased lysis was observed when using 5 and 10 μM nbHER2, while a sharp reduction of NK lytic activity was observed at the higher concentration of 20 μM nbHER2Tyr.
References
ARTICLE SECTIONSThis article references 34 other publications.
- 1El Muslemany, K. M.; Twite, A. A.; ElSohly, A. M.; Obermeyer, A. C.; Mathies, R. A.; Francis, M. B. Photoactivated Bioconjugation Betweenortho-Azidophenols and Anilines: A Facile Approach to Biomolecular Photopatterning. J. Am. Chem. Soc. 2014, 136, 12600– 12606, DOI: 10.1021/ja503056x[ACS Full Text
], [CAS], Google Scholar
1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsVGht7bL&md5=4c5e84d4074baace09004685a2f0b852Photoactivated Bioconjugation Between ortho-Azidophenols and Anilines: A Facile Approach to Biomolecular PhotopatterningEl Muslemany, Kareem M.; Twite, Amy A.; El Sohly, Adel M.; Obermeyer, Allie C.; Mathies, Richard A.; Francis, Matthew B.Journal of the American Chemical Society (2014), 136 (36), 12600-12606CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Methods for the surface patterning of small mols. and biomols. can yield useful platforms for drug screening, synthetic biol. applications, diagnostics, and the immobilization of live cells. However, new techniques are needed to achieve the ease, feature sizes, reliability, and patterning speed necessary for widespread adoption. Herein, the authors report an easily accessible and operationally simple photoinitiated reaction that can achieve patterned bioconjugation in a highly chemoselective manner. The reaction involves the photolysis of 2-azidophenols to generate iminoquinone intermediates that couple rapidly to aniline groups. The authors demonstrate the broad functional group compatibility of this reaction for the modification of proteins, polymers, oligonucleotides, peptides, and small mols. As a specific application, the reaction was adapted for the photolithog. patterning of azidophenol DNA on aniline glass substrates. The presence of the DNA was confirmed by the ability of the surface to capture living cells bearing the sequence complement on their cell walls or cytoplasmic membranes. Compared to other light-based DNA patterning methods, this reaction offers higher speed and does not require the use of a photoresist or other blocking material. - 2Furst, A. L.; Smith, M. J.; Francis, M. B. Direct Electrochemical Bioconjugation on Metal Surfaces. J. Am. Chem. Soc. 2017, 139, 12610– 12616, DOI: 10.1021/jacs.7b06385[ACS Full Text
], [CAS], Google Scholar
2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsVSlu7%252FI&md5=919d942661541624c0a425c5d6d8c3a4Direct Electrochemical Bioconjugation on Metal SurfacesFurst, Ariel L.; Smith, Matthew J.; Francis, Matthew B.Journal of the American Chemical Society (2017), 139 (36), 12610-12616CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)DNA has unique capabilities for mol. recognition and self-assembly, which have fostered its widespread incorporation into devices that are useful in science and medicine. Many of these platforms rely on thiol groups to tether DNA to gold surfaces, but this method is hindered by a lack of control over monolayer d. and by secondary interactions between the nucleotide bases and the metal. In this work, the authors report an electrochem. activated bioconjugation reaction as a mild, reagent-free strategy to attach oligonucleotides to gold surfaces. Aniline-modified DNA was coupled to catechol-coated electrodes that were oxidized to o-quinones using an applied potential. High levels of coupling could be achieved in minutes. By changing the reaction time and the underlying catechol content, the final DNA surface coverage could be specified. The advantages of this method were demonstrated through the electrochem. detection of the endocrine disruptor bisphenol A, as well as the capture of living nonadherent cells on electrode surfaces by DNA hybridization. This method not only improves the attachment of DNA to metal surfaces but also represents a new direction for the site-specific attachment of biomols. to device platforms. - 3Twite, A. A.; Hsiao, S. C.; Onoe, H.; Mathies, R. A.; Francis, M. B. Direct Attachment of Microbial Organisms to Material Surfaces Through Sequence-Specific DNA Hybridization. Adv. Mater. 2012, 24, 2380– 2385, DOI: 10.1002/adma.201104336[Crossref], [PubMed], [CAS], Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XktFShtLg%253D&md5=ea091e8667c10160fd77d77992e25ea6Direct attachment of microbial organisms to material surfaces through sequence-specific DNA hybridizationTwite, Amy A.; Hsiao, Sonny C.; Onoe, Hiroaki; Mathies, Richard A.; Francis, Matthew B.Advanced Materials (Weinheim, Germany) (2012), 24 (18), 2380-2385CODEN: ADVMEW; ISSN:0935-9648. (Wiley-VCH Verlag GmbH & Co. KGaA)General methods are described for the immobilization of microorganisms on patterned surfaces. A new oxidative strategy for the introduction of nucleic acid strands on microbial cell surfaces results in their strong, efficient, and specific binding to surfaces displaying the sequence complement without apparent losses in cell viability. The two-step method to attach ssDNA to these cell types comprises the initial oxidn. of the 1,2-diols in the carbohydrates with sodium periodate, followed by condensation of the resulting aldehyde groups with 5'-hydrazide-terminated ssDNA to form hydrazones. Hydrazone formation was facilitated by the addn. of aniline (10-25 mM) to the reaction mixt. as a catalyst. After optimizing the cell modification scheme for ssDNA, the ability of the strands to mediate surface adhesion was demonstrated with glass slides prepd. with circular patterns of complementary ssDNA using a previously reported reductive animation procedure. Particular advantages of this approach include its high binding efficiency, its minimal impact on cell viability, and its ability to create complex patterns consisting of many different organisms.
- 4Gartner, Z. J.; Bertozzi, C. R. Programmed Assembly of 3-Dimensional Microtissues with Defined Cellular Connectivity. Proc. National Acad. Sci. 2009, 106, 4606– 4610, DOI: 10.1073/pnas.0900717106[Crossref], [PubMed], [CAS], Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXktFOgsLg%253D&md5=00ecc4ecb82b8359ed33a0ff4d418ff6Programmed assembly of 3-dimensional microtissues with defined cellular connectivityGartner, Zev J.; Bertozzi, Carolyn R.Proceedings of the National Academy of Sciences of the United States of America (2009), 106 (12), 4606-4610CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Multicellular organs comprise differentiated cell types with discrete yet interdependent functions. The cells' spatial arrangements and interconnectivities, both crit. elements of higher-order function, derive from complex developmental programs in vivo and are often difficult or impossible to emulate in vitro. Here, we report the bottom-up synthesis of microtissues composed of multiple cell types with programmed connectivity. We functionalized cells with short oligonucleotides to impart specific adhesive properties. Hybridization of complementary DNA sequences enabled the assembly of multicellular structures with defined cell-cell contacts. We demonstrated that the kinetic parameters of the assembly process depend on DNA sequence complexity, d., and total cell concn. Thus, cell assembly can be highly controlled, enabling the design of microtissues with defined cell compn. and stoichiometry. We used this strategy to construct a paracrine signaling network in isolated 3-dimensional microtissues.
- 5Li, D.; Li, X.; Zhou, W.-L.; Huang, Y.; Liang, X.; Jiang, L.; Yang, X.; Sun, J.; Li, Z.; Han, W.-D.; Wang, W. Genetically Engineered T Cells for Cancer Immunotherapy. Signal Transduct. Target Ther. 2019, 4, 35, DOI: 10.1038/s41392-019-0070-9[Crossref], [PubMed], [CAS], Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MjgtlSqsQ%253D%253D&md5=bbfa61151e99ab8538ac41cf89867e40Genetically engineered T cells for cancer immunotherapyLi Dan; Li Xue; Zhou Wei-Lin; Huang Yong; Liang Xiao; Jiang Lin; Yang Xiao; Wang Wei; Liang Xiao; Sun Jie; Sun Jie; Li Zonghai; Li Zonghai; Han Wei-DongSignal transduction and targeted therapy (2019), 4 (), 35 ISSN:.T cells in the immune system protect the human body from infection by pathogens and clear mutant cells through specific recognition by T cell receptors (TCRs). Cancer immunotherapy, by relying on this basic recognition method, boosts the antitumor efficacy of T cells by unleashing the inhibition of immune checkpoints and expands adaptive immunity by facilitating the adoptive transfer of genetically engineered T cells. T cells genetically equipped with chimeric antigen receptors (CARs) or TCRs have shown remarkable effectiveness in treating some hematological malignancies, although the efficacy of engineered T cells in treating solid tumors is far from satisfactory. In this review, we summarize the development of genetically engineered T cells, outline the most recent studies investigating genetically engineered T cells for cancer immunotherapy, and discuss strategies for improving the performance of these T cells in fighting cancers.
- 6Abbina, S.; Siren, E. M. J.; Moon, H.; Kizhakkedathu, J. N. Surface Engineering for Cell-Based Therapies: Techniques for Manipulating Mammalian Cell Surfaces. ACS Biomater. Sci. Eng. 2018, 4, 3658– 3677, DOI: 10.1021/acsbiomaterials.7b00514[ACS Full Text
], [CAS], Google Scholar
6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsVKmtbnN&md5=d38c4c7e8650957daec8026a52084dc3Surface Engineering for Cell-Based Therapies: Techniques for Manipulating Mammalian Cell SurfacesAbbina, Srinivas; Siren, Erika M. J.; Moon, Haisle; Kizhakkedathu, Jayachandran N.ACS Biomaterials Science & Engineering (2018), 4 (11), 3658-3677CODEN: ABSEBA; ISSN:2373-9878. (American Chemical Society)A review. The introduction of cell-based therapies has provided new and unique strategies to treat many diseases and disorders including the recent approval of CAR-T cell therapy for the leukemia. Cell surface engineering is a methodol. in which the cell surface is tailored to modulate cellular function and interactions. In addn. to genetic engineering of cell surface proteins, a wide array of robust, innovative and elegant approaches have been developed to selectivity target the cell surface. In this review, we will introduce the leading strategies currently used in cell surface engineering including broadly reactive chem. ligations and phys. assocns. as well as more controlled approaches as demonstrated in genetic, enzymic and metabolic engineering. Prominent applications of these strategies for cell-based therapies will be highlighted including targeted cell death, controlling stem cell fate, immunoevasion, blood transfusion and the delivery of cells to target tissues. Advances will be focused specifically on cells which are the most promising in generating cell-based therapeutics including red blood cells, white blood cells (lymphocytes, macrophages), stem cells (multipotent and pluripotent), islet cells, cancer cells and endothelial cells. - 7Stephan, M. T.; Irvine, D. J. Enhancing Cell Therapies from the Outside in: Cell Surface Engineering Using Synthetic Nanomaterials. Nano Today. 2011, 6, 309– 325, DOI: 10.1016/j.nantod.2011.04.001[Crossref], [PubMed], [CAS], Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXpvVWqtbc%253D&md5=2a9955bfe0a5a1d915a7d58063464149Enhancing cell therapies from the outside in: cell surface engineering using synthetic nanomaterialsStephan, Matthias T.; Irvine, Darrell J.Nano Today (2011), 6 (3), 309-325CODEN: NTAOCG; ISSN:1748-0132. (Elsevier Ltd.)A review. Therapeutic treatments based on the injection of living cells are in clin. use and preclin. development for diseases ranging from cancer to cardiovascular disease to diabetes. To enhance the function of therapeutic cells, a variety of chem. and materials science strategies are being developed that engineer the surface of therapeutic cells with new mols., artificial receptors, and multifunctional nanomaterials, synthetically endowing donor cells with new properties and functions. These approaches offer a powerful complement to traditional genetic engineering strategies for enhancing the function of living cells.
- 8Shearer, R. F.; Saunders, D. N. Experimental Design for Stable Genetic Manipulation in Mammalian Cell Lines: Lentivirus and Alternatives. Genes Cells. 2015, 20, 1– 10, DOI: 10.1111/gtc.12183[Crossref], [PubMed], [CAS], Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXislSgsA%253D%253D&md5=061a2b69ebbb5d5f10a0ad17ce935c07Experimental design for stable genetic manipulation in mammalian cell lines: lentivirus and alternativesShearer, Robert F.; Saunders, Darren N.Genes to Cells (2015), 20 (1), 1-10CODEN: GECEFL; ISSN:1356-9597. (Wiley-Blackwell)A review. The use of third-generation lentiviral vectors is now commonplace in most areas of basic biol. These systems provide a fast, efficient means for modulating gene expression, but exptl. design needs to be carefully considered to minimize potential artifacts arising from off-target effects and other confounding factors. This review offers a starting point for those new to lentiviral-based vector systems, addressing the main issues involved with the use of lentiviral systems in vitro and outlines considerations which should be taken into account during exptl. design. Factors such as selecting an appropriate system and controls, and practical titrn. of viral transduction are important considerations for exptl. design. We also briefly describe some of the more recent advances in genome editing technol. TALENs and CRISPRs offer an alternative to lentivirus, providing endogenous gene editing with reduced off-target effects often at the expense of efficiency.
- 9Kohn, D. B.; Sadelain, M.; Glorioso, J. C. Occurrence of Leukaemia Following Gene Therapy of X-Linked SCID. Nat. Rev. Cancer. 2003, 3, 477– 488, DOI: 10.1038/nrc1122[Crossref], [PubMed], [CAS], Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXltVSqtrw%253D&md5=a51e02e95ee60bd85e3a30a4767d8b98Occurrence of leukemia following gene therapy of X-linked SCIDKohn, Donald B.; Sadelain, Michel; Glorioso, Joseph C.Nature Reviews Cancer (2003), 3 (7), 477-488CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)A review. Recombinant viral vectors have allowed gene transfer to be developed as a promising approach to the treatment of genetic diseases. Recently, gene therapy of children with X-linked severe combined immune deficiency resulted in impressive levels of immune reconstitution - a triumph that was later overshadowed by the development of leukemia in 2 patients. What were the causes of this cancer, and how can the therapeutic benefits of gene therapy be achieved while minimizing risk to the patient.
- 10Prescher, J. A.; Dube, D. H.; Bertozzi, C. R. Chemical Remodelling of Cell Surfaces in Living Animals. Nature. 2004, 430, 873– 877, DOI: 10.1038/nature02791[Crossref], [PubMed], [CAS], Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXmslCnu70%253D&md5=add5fdac7607d716a5564137ca6c06baChemical remodelling of cell surfaces in living animalsPrescher, Jennifer A.; Dube, Danielle H.; Bertozzi, Carolyn R.Nature (London, United Kingdom) (2004), 430 (7002), 873-877CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Cell surfaces are endowed with biol. functionality designed to mediate extracellular communication. The cell-surface repertoire can be expanded to include abiotic functionality through the biosynthetic introduction of unnatural sugars into cellular glycans, a process termed metabolic oligosaccharide engineering. This technique has been exploited in fundamental studies of glycan-dependent cell-cell and virus-cell interactions and also provides an avenue for the chem. remodelling of living cells. Unique chem. functional groups can be delivered to cell-surface glycans by metab. of the corresponding unnatural precursor sugars. These functional groups can then undergo covalent reaction with exogenous agents bearing complementary functionality. The exquisite chem. selectivity required of this process is supplied by the Staudinger ligation of azides and phosphines, a reaction that has been performed on cultured cells without detriment to their physiol. Here we demonstrate that the Staudinger ligation can be executed in living animals, enabling the chem. modification of cells within their native environment. The ability to tag cell-surface glycans in vivo may enable therapeutic targeting and non-invasive imaging of changes in glycosylation during disease progression.
- 11Baskin, J. M.; Prescher, J. A.; Laughlin, S. T.; Agard, N. J.; Chang, P. V.; Miller, I. A.; Lo, A.; Codelli, J. A.; Bertozzi, C. R. Copper-Free Click Chemistry for Dynamic in Vivo Imaging. Proc. National Acad. Sci. 2007, 104, 16793– 16797, DOI: 10.1073/pnas.0707090104[Crossref], [PubMed], [CAS], Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXht1KgtLvN&md5=7ea429c09b86509ff7f3960a95f11276Copper-free click chemistry for dynamic in vivo imagingBaskin, Jeremy M.; Prescher, Jennifer A.; Laughlin, Scott T.; Agard, Nicholas J.; Chang, Pamela V.; Miller, Isaac A.; Lo, Anderson; Codelli, Julian A.; Bertozzi, Carolyn R.Proceedings of the National Academy of Sciences of the United States of America (2007), 104 (43), 16793-16797CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Dynamic imaging of proteins in live cells is routinely performed by using genetically encoded reporters, an approach that cannot be extended to other classes of biomols. such as glycans and lipids. Here, the authors report a Cu-free variant of click chem. that can label these biomols. rapidly and selectively in living systems, overcoming the intrinsic toxicity of the canonical Cu-catalyzed reaction. The crit. reagent, a substituted cyclooctyne, possesses ring strain and electron-withdrawing fluorine substituents that together promote the [3+2] dipolar cycloaddn. with azides installed metabolically into biomols. This Cu-free click reaction possesses comparable kinetics to the Cu-catalyzed reaction and proceeds within minutes on live cells with no apparent toxicity. With this technique, the authors studied the dynamics of glycan trafficking and identified a population of sialoglycoconjugates with unexpectedly rapid internalization kinetics.
- 12Saxon, E.; Bertozzi, C. R. Cell Surface Engineering by a Modified Staudinger Reaction. Science. 2000, 287, 2007– 2010, DOI: 10.1126/science.287.5460.2007[Crossref], [PubMed], [CAS], Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXhvF2ru7g%253D&md5=91685aaba684aa4dfee86f44fef09886Cell surface engineering by a modified Staudinger reactionSaxon, Eliana; Bertozzi, Carolyn R.Science (Washington, D. C.) (2000), 287 (5460), 2007-2010CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Selective chem. reactions enacted within a cellular environment can be powerful tools for elucidating biol. processes or engineering novel interactions. A chem. transformation that permits the selective formation of covalent adducts among richly functionalized biopolymers within a cellular context is presented. A ligation modeled after the Staudinger reaction forms an amide bond by coupling of an azide and a specifically engineered triarylphosphine. Both reactive partners are abiotic and chem. orthogonal to native cellular components. Azides installed within cell surface glycoconjugates by metab. of a synthetic azidosugar were reacted with a biotinylated triarylphosphine to produce stable cell-surface adducts. The tremendous selectivity of the transformation should permit its execution within a cell's interior, offering new possibilities for probing intracellular interactions.
- 13Wang, X.; Lang, S.; Tian, Y.; Zhang, J.; Yan, X.; Fang, Z.; Weng, J.; Lu, N.; Wu, X.; Li, T.; Cao, H.; Li, Z.; Huang, X. Glycoengineering of Natural Killer Cells with CD22 Ligands for Enhanced Anticancer Immunotherapy. ACS Central Sci. 2020, 6, 382– 389, DOI: 10.1021/acscentsci.9b00956[ACS Full Text
], [CAS], Google Scholar
13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkt1yks70%253D&md5=5f6f36cf07a6f0428feb33487bfe6807Glycoengineering of Natural Killer Cells with CD22 Ligands for Enhanced Anticancer ImmunotherapyWang, Xianwu; Lang, Shuyao; Tian, Yunpeng; Zhang, Jianghong; Yan, Xu; Fang, Zhihong; Weng, Jian; Lu, Na; Wu, Xuanjun; Li, Tianlu; Cao, Hongzhi; Li, Zhu; Huang, XuefeiACS Central Science (2020), 6 (3), 382-389CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)Adoptive transfer of immune cells is being actively pursued for cancer treatment. Natural killer (NK) cells, a class of cytotoxic immune cells, generally lack inherent selectivities toward cancer. To bestow tumor-targeting abilities and enhance anticancer efficacy, a new strategy is established to glycoengineer NK cells. Carbohydrate-based ligands for CD22, a marker for B cell lymphoma, are introduced onto NK cells through either metabolic engineering or glyco-polymer insertion. Such NK cells exhibited greatly enhanced cytotoxicities toward CD22+ lymphoma cells in a CD22-dependent manner. Importantly, both CD22+ lymphoma cell lines and primary lymphoma cells from human cancer patients can be effectively killed by the engineered NK cells. Furthermore, glycoengineered NK cells provided significant protection to tumor-bearing mice. Thus, NK cell glycoengineering is an exciting new approach for cancer treatment complementing the current immune cell genetic engineering strategy. Sweetening natural killer cells enhanced their tumor-killing abilities in vitro and reduced tumor growth in vivo. - 14Frank, M. J.; Olsson, N.; Huang, A.; Tang, S.-W.; Negrin, R. S.; Elias, J. E.; Meyer, E. H. A Novel Antibody-Cell Conjugation Method to Enhance and Characterize Cytokine-Induced Killer Cells. Cytotherapy. 2020, 22, 135– 143, DOI: 10.1016/j.jcyt.2020.01.003[Crossref], [PubMed], [CAS], Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXltFykt7w%253D&md5=c61c8f677e77dd3c8edd1a4c3e24f9d4A novel antibody-cell conjugation method to enhance and characterize cytokine-induced killer cellsFrank, Matthew J.; Olsson, Niclas; Huang, Andy; Tang, Sai-Wen; Negrin, Robert S.; Elias, Joshua E.; Meyer, Everett H.Cytotherapy (2020), 22 (3), 135-143CODEN: CYTRF3; ISSN:1465-3249. (Elsevier Inc.)Cytokine-induced killer (CIK) cells are an ex vivo-expanded cellular therapy product with potent anti-tumor activity in a subset of patients with solid and hematol. malignancies. We hypothesize that directing CIK cells to a specific tumor antigen will enhance CIK cell anti-tumor cytotoxicity. We present a newly developed method for affixing antibodies directly to cell surface proteins. First, we evaluated the anti-tumor potential of CIK cells after affixing tumor-antigen targeting monoclonal antibodies. Second, we evaluated whether this antibody-conjugation method can profile the surface proteome of CIK cells. We demonstrated that affixing rituximab or daratumumab to CIK cells enhances cytotoxic killing of multiple lymphoma cell lines in vitro. These armed CIK cells exhibited enhanced intracellular signaling after engaging tumor targets. Cell surface proteome profiling suggested mechanisms by which antibody-armed CIK cells concurrently activated multiple surface proteins, leading to enhanced cytolytic activity. Our surface proteome anal. indicated that CIK cells display enhanced protein signatures indicative of immune responses, cellular activation and leukocyte migration. Here, we characterize the cell surface proteome of CIK cells using a novel methodol. that can be rapidly applied to other cell types. Without genetic modification CIK cells can be rapidly armed with monoclonal antibodies, which endows them with high specificity to kill tumor targets.
- 15Li, H.-K.; Hsiao, C.-W.; Yang, S.-H.; Yang, H.-P.; Wu, T.-S.; Lee, C.-Y.; Lin, Y.-L.; Pan, J.; Cheng, Z.-F.; Lai, Y.-D.; Hsiao, S.-C.; Tang, S.-W. A Novel Off-the-Shelf Trastuzumab-Armed NK Cell Therapy (ACE1702) Using Antibody-Cell-Conjugation Technology. Cancers. 2021, 13 (11), 2724, DOI: 10.3390/cancers13112724[Crossref], [PubMed], [CAS], Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXislehs7zO&md5=3945f97c94b8dbc432efd61eb8dfd07eA Novel off-the-Shelf Trastuzumab-Armed NK Cell Therapy (ACE1702) Using Antibody-Cell-Conjugation TechnologyLi, Hao-Kang; Hsiao, Ching-Wen; Yang, Sen-Han; Yang, Hsiu-Ping; Wu, Tai-Sheng; Lee, Chia-Yun; Lin, Yan-Liang; Pan, Janet; Cheng, Zih-Fei; Lai, Yan-Da; Hsiao, Shih-Chia; Tang, Sai-WenCancers (2021), 13 (11), 2724CODEN: CANCCT; ISSN:2072-6694. (MDPI AG)Simple Summary: Chimeric antigen receptor T cell therapy has shown its potency against hematol. malignancies in autologous settings but also limited success against solid tumors with severe adverse events, including fatal cases of cytokine releasing syndrome. The aim of this research is to develop a novel off-the-shelf natural killer cell therapy against HER2-expressing cancers using Antibody-Cell Conjugation (ACC) technol. and the endogenous CD16-expressing oNK cell line. ACE1702, trastuzumab-armed oNK cells with γ irradn. and cryopreservation, present superior in vitro and in vivo potency against HER2-expressing cancer cells and shows no tumorigenic potential, indicating the clin. application fighting HER2-expressing solid tumors. These findings suggest that ACC technol. can be applied to allogeneic immune cells to provide off-the-shelf therapies for cancer patients. Natural killer (NK) cells harbor efficient cytotoxicity against tumor cells without causing life-threatening cytokine release syndrome (CRS) or graft-vs.-host disease (GvHD). When compared to chimeric antigen receptor (CAR) technol., Antibody-Cell Conjugation (ACC) technol. has been developed to provide an efficient platform to arm immune cells with cancer-targeting antibodies to recognize and attack cancer cells. Recently, we established an endogenous CD16-expressing oNK cell line (oNK) with a favorable expression pattern of NK activation/inhibitory receptors. In this study, we applied ACC platform to conjugate oNK with trastuzumab and an anti-human epidermal growth factor receptor 2 (HER2) antibody. Trastuzumab-conjugated oNK, ACE-oNK-HER2, executed in vitro and in vivo cytotoxicity against HER2-expressing cancer cells and secretion of IFNγ. The irradiated and cryopreserved ACE-oNK-HER2, designated as ACE1702, retained superior HER2-specific in vitro and in vivo potency with no tumorigenic potential. In conclusion, this study provides the evidence to support the potential clin. application of ACE1702 as a novel off-the-shelf NK cell therapy against HER2-expressing solid tumors.
- 16Pishesha, N.; Bilate, A. M.; Wibowo, M. C.; Huang, N.-J.; Li, Z.; Deshycka, R.; Bousbaine, D.; Li, H.; Patterson, H. C.; Dougan, S. K.; Maruyama, T.; Lodish, H. F.; Ploegh, H. L. Engineered Erythrocytes Covalently Linked to Antigenic Peptides Can Protect against Autoimmune Disease. Proc. National Acad. Sci. 2017, 114, 3157– 3162, DOI: 10.1073/pnas.1701746114[Crossref], [PubMed], [CAS], Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjvVyjs7w%253D&md5=6a129adca06524e386953054bc3dd62eEngineered erythrocytes covalently linked to antigenic peptides can protect against autoimmune diseasePishesha, Novalia; Bilate, Angelina M.; Wibowo, Marsha C.; Huang, Nai-Jia; Li, Zeyang; Dhesycka, Rhogerry; Bousbaine, Djenet; Li, Hojun; Patterson, Heide C.; Dougan, Stephanie K.; Maruyama, Takeshi; Lodish, Harvey F.; Ploegh, Hidde L.Proceedings of the National Academy of Sciences of the United States of America (2017), 114 (12), 3157-3162CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Current therapies for autoimmune diseases rely on traditional immunosuppressive medications that expose patients to an increased risk of opportunistic infections and other complications. Immunoregulatory interventions that act prophylactically or therapeutically to induce antigen-specific tolerance might overcome these obstacles. Here the authors use the transpeptidase sortase to covalently attach disease-assocd. autoantigens to genetically engineered and to unmodified red blood cells as a means of inducing antigen-specific tolerance. This approach blunts the contribution to immunity of major subsets of immune effector cells (B cells, CD4+ and CD8+ T cells) in an antigen-specific manner. Transfusion of red blood cells expressing self-antigen epitopes can alleviate and even prevent signs of disease in exptl. autoimmune encephalomyelitis, as well as maintain normoglycemia in a mouse model of type 1 diabetes.
- 17Shi, J.; Kundrat, L.; Pishesha, N.; Bilate, A.; Theile, C.; Maruyama, T.; Dougan, S. K.; Ploegh, H. L.; Lodish, H. F. Engineered Red Blood Cells as Carriers for Systemic Delivery of a Wide Array of Functional Probes. Proc. National Acad. Sci. 2014, 111, 10131– 10136, DOI: 10.1073/pnas.1409861111[Crossref], [PubMed], [CAS], Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtVOit7vL&md5=0c8fc0c8a427682d0435231268bd71ddEngineered red blood cells as carriers for systemic delivery of a wide array of functional probesShi, Jiahai; Kundrat, Lenka; Pishesha, Novalia; Bilate, Angelina; Theile, Chris; Maruyama, Takeshi; Dougan, Stephanie K.; Ploegh, Hidde L.; Lodish, Harvey F.Proceedings of the National Academy of Sciences of the United States of America (2014), 111 (28), 10131-10136CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)We developed modified RBCs to serve as carriers for systemic delivery of a wide array of payloads. These RBCs contain modified proteins on their plasma membrane, which can be labeled in a sortase-catalyzed reaction under native conditions without inflicting damage to the target membrane or cell. Sortase accommodates a wide range of natural and synthetic payloads that allow modification of RBCs with substituents that cannot be encoded genetically. As proof of principle, we demonstrate site-specific conjugation of biotin to in vitro-differentiated mouse erythroblasts as well as to mature mouse RBCs. Thus modified, RBCs remain in the bloodstream for up to 28 d. A single domain antibody attached enzymically to RBCs enables them to bind specifically to target cells that express the antibody target. We extend these expts. to human RBCs and demonstrate efficient sortase-mediated labeling of in vitro-differentiated human reticulocytes.
- 18Harmand, T. J.; Pishesha, N.; Rehm, F. B. H.; Ma, W.; Pinney, W. B.; Xie, Y. J.; Ploegh, H. L. Asparaginyl Ligase-Catalyzed One-Step Cell Surface Modification of Red Blood Cells. ACS Chem. Biol. 2021, 16, 1201– 1207, DOI: 10.1021/acschembio.1c00216[ACS Full Text
], [CAS], Google Scholar
18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtlSmu7jI&md5=cdf20ff3ecfc82c4b1b50bb019c90306Asparaginyl Ligase-Catalyzed One-Step Cell Surface Modification of Red Blood CellsHarmand, Thibault J.; Pishesha, Novalia; Rehm, Fabian B. H.; Ma, Weiyi; Pinney, William B.; Xie, Yushu J.; Ploegh, Hidde L.ACS Chemical Biology (2021), 16 (7), 1201-1207CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Red blood cells (RBCs) can serve as vascular carriers for drugs, proteins, peptides, and nanoparticles. Human RBCs remain in the circulation for ∼120 days, are biocompatible, and are immunol. largely inert. RBCs are cleared by the reticuloendothelial system and can induce immune tolerance to foreign components attached to the RBC surface. RBC conjugates have been pursued in clin. trials to treat cancers and autoimmune diseases and to correct genetic disorders. Still, most methods used to modify RBCs require multiple steps, are resource-intensive and time-consuming, and increase the risk of inflicting damage to the RBCs. Here, we describe direct conjugation of peptides and proteins onto the surface of RBCs in a single step, catalyzed by a highly efficient, recombinant asparaginyl ligase under mild, physiol. conditions. In mice, the modified RBCs remain intact in the circulation, display a normal circulatory half-life, and retain their immune tolerance-inducing properties, as shown for protection against an accelerated model for type 1 diabetes. We conjugated different nanobodies to RBCs with retention of their binding properties, and these modified RBCs can target cancer cells in vitro. This approach provides an appealing alternative to current methods of RBC engineering. It provides ready access to more complex RBC constructs and highlights the general utility of asparaginyl ligases for the modification of native cell surfaces. - 19Li, J.; Chen, M.; Liu, Z.; Zhang, L.; Felding, B. H.; Moremen, K. W.; Lauvau, G.; Abadier, M.; Ley, K.; Wu, P. A Single-Step Chemoenzymatic Reaction for the Construction of Antibody–Cell Conjugates. ACS Central Sci. 2018, 4, 1633– 1641, DOI: 10.1021/acscentsci.8b00552[ACS Full Text
], [CAS], Google Scholar
19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisVaqsrbP&md5=59cb733e3db9f5b9ffb711451afd68b4A Single-Step Chemoenzymatic Reaction for the Construction of Antibody-Cell ConjugatesLi, Jie; Chen, Mingkuan; Liu, Zilei; Zhang, Linda; Felding, Brunie H.; Moremen, Kelley W.; Lauvau, Gregoire; Abadier, Michael; Ley, Klaus; Wu, PengACS Central Science (2018), 4 (12), 1633-1641CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)Employing live cells as therapeutics is a direction of future drug discovery. An easy and robust method to modify the surfaces of cells directly to incorporate novel functionalities is highly desirable. However, genetic methods for cell-surface engineering are laborious and limited by low efficiency for primary cell modification. Here we report a chemoenzymic approach that exploits a fucosyltransferase to transfer bio-macromols., such as an IgG antibody (MW∼ 150 kDa), to the glycocalyx on the surfaces of live cells when the antibody is conjugated to the enzyme's natural donor substrate GDP-Fucose. Requiring no genetic modification, this method is fast and biocompatible with little interference to cells' endogenous functions. We applied this method to construct two antibody-cell conjugates (ACCs) using both cell lines and primary cells, and the modified cells exhibited specific tumor targeting and resistance to inhibitory signals produced by tumor cells, resp. Remarkably, Herceptin-NK-92MI conjugates, a natural killer cell line modified with Herceptin, exhibit enhanced activities to induce the lysis of HER2+ cancer cells both ex vivo and in a human tumor xenograft model. Given the unprecedented substrate tolerance of the fucosyltransferase, this chemoenzymic method offers a general approach to engineer cells as research tools and for therapeutic applications. - 20Maza, J. C.; Bader, D. L. V.; Xiao, L.; Marmelstein, A. M.; Brauer, D. D.; ElSohly, A. M.; Smith, M. J.; Krska, S. W.; Parish, C. A.; Francis, M. B. Enzymatic Modification of N-Terminal Proline Residues Using Phenol Derivatives. J. Am. Chem. Soc. 2019, 141, 3885– 3892, DOI: 10.1021/jacs.8b10845[ACS Full Text
], [CAS], Google Scholar
20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXisFyitbc%253D&md5=8b203591be06bb432717258ea0452ca3Enzymatic Modification of N-Terminal Proline Residues Using Phenol DerivativesMaza, Johnathan C.; Bader, Daniel L. V.; Xiao, Lifeng; Marmelstein, Alan M.; Brauer, Daniel D.; El Sohly, Adel M.; Smith, Matthew J.; Krska, Shane W.; Parish, Craig A.; Francis, Matthew B.Journal of the American Chemical Society (2019), 141 (9), 3885-3892CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)A convenient enzymic strategy is reported for the modification of proline residues in the N-terminal positions of proteins. Using a tyrosinase enzyme isolated from Agaricus bisporus (abTYR), phenols and catechols are oxidized to highly reactive o-quinone intermediates that then couple to N-terminal proline residues in high yield. Key advantages of this bioconjugation method include (1) the use of air-stable precursors that can be prepd. on large scale if needed, (2) mild reaction conditions, including low temps., (3) the targeting of native functional groups that can be introduced readily on most proteins, and (4) the use of mol. oxygen as the sole oxidant. This coupling strategy was successfully demonstrated for the attachment of a variety of phenol-derivatized cargo mols. to a series of protein substrates, including self-assembled viral capsids, enzymes, and a chitin binding domain (CBD). The ability of the CBD to bind to the surfaces of yeast cells was found to be unperturbed by this modification reaction. - 21Lobba, M. J.; Fellmann, C.; Marmelstein, A. M.; Maza, J. C.; Kissman, E. N.; Robinson, S. A.; Staahl, B. T.; Urnes, C.; Lew, R. J.; Mogilevsky, C. S.; Doudna, J. A.; Francis, M. B. Site-Specific Bioconjugation through Enzyme-Catalyzed Tyrosine–Cysteine Bond Formation. ACS Central Sci. 2020, 6, 1564– 1571, DOI: 10.1021/acscentsci.0c00940[ACS Full Text
], [CAS], Google Scholar
21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs1eltL3P&md5=a0fe6337ef9daa4caa637a7ba54f9d68Site-Specific Bioconjugation through Enzyme-Catalyzed Tyrosine-Cysteine Bond FormationLobba, Marco J.; Fellmann, Christof; Marmelstein, Alan M.; Maza, Johnathan C.; Kissman, Elijah N.; Robinson, Stephanie A.; Staahl, Brett T.; Urnes, Cole; Lew, Rachel J.; Mogilevsky, Casey S.; Doudna, Jennifer A.; Francis, Matthew B.ACS Central Science (2020), 6 (9), 1564-1571CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)The synthesis of protein-protein and protein-peptide conjugates is an important capability for producing vaccines, immunotherapeutics, and targeted delivery agents. Herein we show that the enzyme tyrosinase is capable of oxidizing exposed tyrosine residues into o-quinones that react rapidly with cysteine residues on target proteins. This coupling reaction occurs under mild aerobic conditions and has the rare ability to join full-size proteins in under 2 h. The utility of the approach is demonstrated for the attachment of cationic peptides to enhance the cellular delivery of CRISPR-Cas9 20-fold and for the coupling of reporter proteins to a cancer-targeting antibody fragment without loss of its cell-specific binding ability. The broad applicability of this technique provides a new building block approach for the synthesis of protein chimeras. Enzymic oxidn. of tyrosine residues followed by reaction with cysteine thiols allows the covalent coupling of proteins and peptides using only native amino acid side chains. - 22Marmelstein, A. M.; Lobba, M. J.; Mogilevsky, C. S.; Maza, J. C.; Brauer, D. D.; Francis, M. B. Tyrosinase-Mediated Oxidative Coupling of Tyrosine Tags on Peptides and Proteins. J. Am. Chem. Soc. 2020, 142, 5078– 5086, DOI: 10.1021/jacs.9b12002[ACS Full Text
], [CAS], Google Scholar
22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXjs12gtLc%253D&md5=e61c218de8ccf478384e3fbbb3407f65Tyrosinase-Mediated Oxidative Coupling of Tyrosine Tags on Peptides and ProteinsMarmelstein, Alan M.; Lobba, Marco J.; Mogilevsky, Casey S.; Maza, Johnathan C.; Brauer, Daniel D.; Francis, Matthew B.Journal of the American Chemical Society (2020), 142 (11), 5078-5086CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Oxidative coupling (OC) through o-quinone intermediates has been established as an efficient and site-selective way to modify protein N-termini and the unnatural amino acid p-aminophenylalanine (paF). Recently, we reported that the tyrosinase-mediated oxidn. of phenol-tagged cargo mols. is a particularly convenient method of generating o-quinones in situ. The coupling partners can be easily prepd. and stored, the reaction takes place under mild conditions (phosphate buffer, pH 6.5, 4 to 23 °C), and dissolved oxygen is the only oxidant required. Here, we show an important extension of this chem. for the activation of tyrosine residues that project into soln. from the N or C-termini of peptide and protein substrates. Generating the o-quinone electrophiles from tyrosine allows greater flexibility in choosing the nucleophilic coupling partner and expands the scope of the reaction to include C-terminal positions. We also introduce a new bacterial tyrosinase enzyme that shows improved activation for some tyrosine substrates. The efficacy of several secondary amines and aniline derivs. was evaluated in the coupling reactions, providing important information for coupling partner design. This strategy was used to modify the C-termini of an antibody scFv construct and of Protein L, a human IgG kappa light chain binding protein. The use of the modified proteins as immunolabeling agents was also demonstrated. - 23Bruins, J. J.; Westphal, A. H.; Albada, B.; Wagner, K.; Bartels, L.; Spits, H.; van Berkel, W. J. H.; van Delft, F. L. Inducible, Site-Specific Protein Labeling by Tyrosine Oxidation–Strain-Promoted (4 + 2) Cycloaddition. Bioconjugate Chem. 2017, 28, 1189– 1193, DOI: 10.1021/acs.bioconjchem.7b00046[ACS Full Text
], [CAS], Google Scholar
23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjs12ntro%253D&md5=058655ebcbdef65719b9a0613dae3d51Inducible, Site-Specific Protein Labeling by Tyrosine Oxidation-Strain-Promoted (4 + 2) CycloadditionBruins, Jorick J.; Westphal, Adrie H.; Albada, Bauke; Wagner, Koen; Bartels, Lina; Spits, Hergen; van Berkel, Willem J. H.; van Delft, Floris L.Bioconjugate Chemistry (2017), 28 (4), 1189-1193CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Genetically encoded tyrosine (Y-tag) can be utilized as a latent anchor for inducible and site-selective conjugation. Upon oxidn. of tyrosine with mushroom tyrosinase, strain-promoted cycloaddn. (SPOCQ) of the resulting 1,2-quinone with various bicyclo[6.1.0]nonyne (BCN) derivs. led to efficient conjugation. The method was applied for fluorophore labeling of laminarinase A and for the site-specific prepn. of an antibody-drug conjugate. - 24Wilton, E. E.; Opyr, M. P.; Kailasam, S.; Kothe, R. F.; Wieden, H.-J. SdAb-DB: The Single Domain Antibody Database. ACS Synth. Biol. 2018, 7, 2480– 2484, DOI: 10.1021/acssynbio.8b00407[ACS Full Text
], [CAS], Google Scholar
24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitFertbbJ&md5=991b0b08079a4f778d8150b902fb010csdAb-DB: The Single Domain Antibody DatabaseWilton, Emily E.; Opyr, Michael P.; Kailasam, Senthilkumar; Kothe, Ronja F.; Wieden, Hans-JoachimACS Synthetic Biology (2018), 7 (11), 2480-2484CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)A review. The Single Domain Antibody Database, or sdAb-DB, (www.sdab-db.ca) is the first freely available repository for single domain antibodies and related classes of proteins. Due to their small size, modular structure, and ease of expression, single domain antibodies (sdAb) have a wide range of applications, including as a rational design tool, and are therefore of great interest for synthetic biologists and bioengineers. However, to enable effective use and sharing of existing sdAbs, including those with engineered functions (e.g., fusions with fluorescent proteins), as well as the rational design and engineering of new sdAbs, it is necessary to have access to sequences and exptl. data. We have therefore developed a publicly available, sdAb-focused database, providing access to manually curated sdAb data from protein databases, published scientific articles, and user submissions. The sdAb-DB is an open-source repository and sharing platform for the sdAb community, providing access to performance data and basic bioinformatic tools for use with previously described and validated sdAbs, as well as for the engineering of new sdAb-based designs and proteins. - 25Kirchhofer, A.; Helma, J.; Schmidthals, K.; Frauer, C.; Cui, S.; Karcher, A.; Pellis, M.; Muyldermans, S.; Casas-Delucchi, C. S.; Cardoso, M. C.; Leonhardt, H.; Hopfner, K.-P.; Rothbauer, U. Modulation of Protein Properties in Living Cells Using Nanobodies. Nat. Struct. Mol. Biol. 2010, 17, 133– 138, DOI: 10.1038/nsmb.1727[Crossref], [PubMed], [CAS], Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsFGjtr3K&md5=893913260f608a5337037fc43891ba24Modulation of protein properties in living cells using nanobodiesKirchhofer, Axel; Helma, Jonas; Schmidthals, Katrin; Frauer, Carina; Cui, Sheng; Karcher, Annette; Pellis, Mireille; Muyldermans, Serge; Casas-Delucchi, Corella S.; Cardoso, M. Cristina; Leonhardt, Heinrich; Hopfner, Karl-Peter; Rothbauer, UlrichNature Structural & Molecular Biology (2010), 17 (1), 133-138CODEN: NSMBCU; ISSN:1545-9993. (Nature Publishing Group)Protein conformation is critically linked to function and often controlled by interactions with regulatory factors. Here we report the selection of camelid-derived single-domain antibodies (nanobodies) that modulate the conformation and spectral properties of the green fluorescent protein (GFP). One nanobody could reversibly reduce GFP fluorescence by a factor of 5, whereas its displacement by a second nanobody caused an increase by a factor of 10. Structural anal. of GFP-nanobody complexes revealed that the two nanobodies induce subtle opposing changes in the chromophore environment, leading to altered absorption properties. Unlike conventional antibodies, the small, stable nanobodies are functional in living cells. Nanobody-induced changes were detected by ratio imaging and used to monitor protein expression and subcellular localization as well as translocation events such as the tamoxifen-induced nuclear localization of estrogen receptor (ER). This work demonstrates that protein conformations can be manipulated and studied with nanobodies in living cells.
- 26Morvan, M. G.; Lanier, L. L. NK Cells and Cancer: You Can Teach Innate Cells New Tricks. Nat. Rev. Cancer. 2016, 16, 7– 19, DOI: 10.1038/nrc.2015.5[Crossref], [PubMed], [CAS], Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXitVOjsbnE&md5=bc3ca6826c04df6147dbe9770b90f420NK cells and cancer: you can teach innate cells new tricksMorvan, Maelig G.; Lanier, Lewis L.Nature Reviews Cancer (2016), 16 (1), 7-19CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)Natural killer (NK) cells are the prototype innate lymphoid cells endowed with potent cytolytic function that provide host defense against microbial infection and tumors. Here, we review evidence for the role of NK cells in immune surveillance against cancer and highlight new therapeutic approaches for targeting NK cells in the treatment of cancer.
- 27Bald, T.; Krummel, M. F.; Smyth, M. J.; Barry, K. C. The NK Cell–Cancer Cycle: Advances and New Challenges in NK Cell–Based Immunotherapies. Nat. Immunol. 2020, 21, 835– 847, DOI: 10.1038/s41590-020-0728-z[Crossref], [PubMed], [CAS], Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVWgt7fK&md5=b47b77228617a9253f7b5dd80319bd23The NK cell-cancer cycle: advances and new challenges in NK cell-based immunotherapiesBald, Tobias; Krummel, Matthew F.; Smyth, Mark J.; Barry, Kevin C.Nature Immunology (2020), 21 (8), 835-847CODEN: NIAMCZ; ISSN:1529-2908. (Nature Research)A review. Abstr.: Natural killer (NK) cells belong to the innate immune system and contribute to protecting the host through killing of infected, foreign, stressed or transformed cells. Addnl., via cellular cross-talk, NK cells orchestrate antitumor immune responses. Hence, significant efforts have been undertaken to exploit the therapeutic properties of NK cells in cancer. Current strategies in preclin. and clin. development include adoptive transfer therapies, direct stimulation, recruitment of NK cells into the tumor microenvironment (TME), blockade of inhibitory receptors that limit NK cell functions, and therapeutic modulation of the TME to enhance antitumor NK cell function. In this Review, we introduce the NK cell-cancer cycle to highlight recent advances in NK cell biol. and to discuss the progress and problems of NK cell-based cancer immunotherapies.
- 28Tam, Y. K.; Maki, G.; Miyagawa, B.; Hennemann, B.; Tonn, T.; Klingemann, H.-G. Characterization of Genetically Altered, Interleukin 2-Independent Natural Killer Cell Lines Suitable for Adoptive Cellular Immunotherapy. Hum. Gene Ther. 1999, 10, 1359– 1373, DOI: 10.1089/10430349950018030[Crossref], [PubMed], [CAS], Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXjsVyqtbs%253D&md5=1c51fc49a1b3c58426dc16e1953f3785Characterization of genetically altered, interleukin 2-independent natural killer cell lines suitable for adoptive cellular immunotherapyTam, Y. K.; Maki, G.; Miyagawa, B.; Hennemann, B.; Tonn, T.; Klingemann, H.-G.Human Gene Therapy (1999), 10 (8), 1359-1373CODEN: HGTHE3; ISSN:1043-0342. (Mary Ann Liebert, Inc.)NK-92 is a highly cytotoxic natural killer (NK) tumor cell line that possesses properties that make it an excellent candidate for adoptive cellular immunotherapy. However, the cytotoxicity of NK cells is dependent on cytokines such as interleukin 2 (IL-2). Although NK-92 cells maintain cytotoxicity for a time after withdrawal of IL-2, clin. use will probably require prolonged treatment with fully activated cells to eliminate disease effectively. The ability to support cytotoxic cells with exogenously administered IL-2 is limited by assocd. toxicity. Therefore, the authors describe the transfection of the IL-2-dependent NK-92 cell line with human IL-2 (hIL-2) cDNA by particle-mediated gene transfer to create two IL-2-independent variants, NK-92MI and NK-92CI, and describe their characterization and comparison with parental cells. Both variants were shown to contain, express, and synthesize the hIL-2 cDNA. IL-2 synthesis was higher in NK-92MI cells compared with NK-92CI cells, with no expression in parental cells. Functionally, the cytotoxicity of all three cell lines was similar and coincubation with IL-2-independent variants did not affect hematopoietic progenitor cells. NK-92MI and NK-92CI cells were more radiosensitive than NK-92 cells, with proliferation inhibited at lower radiation doses and increased morality and decreased cytotoxicity compared with parental cells. Data presented here show that the authors have created by particle-mediated gene transfer two IL-2-independent variants of NK-92 that are identical to parental cells in virtually all respects, including high cytotoxic activity. The nonviral transfection of these cells makes them suitable for clin. applications. These IL-2-independent cells should allow prolonged treatment with fully active natural killer cells without the need for exogenous IL-2 support.
- 29Suck, G.; Odendahl, M.; Nowakowska, P.; Seidl, C.; Wels, W. S.; Klingemann, H. G.; Tonn, T. NK-92: An ‘off-the-Shelf Therapeutic’ for Adoptive Natural Killer Cell-Based Cancer Immunotherapy. Cancer Immunol. Immunother. 2016, 65, 485– 492, DOI: 10.1007/s00262-015-1761-x[Crossref], [PubMed], [CAS], Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvVeqsb7L&md5=2ba14aed3365a23ad62211242c03a2aaNK-92: an 'off-the-shelf therapeutic' for adoptive natural killer cell-based cancer immunotherapySuck, Garnet; Odendahl, Marcus; Nowakowska, Paulina; Seidl, Christian; Wels, Winfried S.; Klingemann, Hans G.; Tonn, TorstenCancer Immunology Immunotherapy (2016), 65 (4), 485-492CODEN: CIIMDN; ISSN:0340-7004. (Springer)Natural killer (NK) cells are increasingly considered as immunotherapeutic agents in particular in the fight against cancers. NK cell therapies are potentially broadly applicable and, different from their T cell counterparts, do not cause graft-vs.-host disease. Efficacy and clin. in vitro or in vivo expansion of primary NK cells will however always remain variable due to individual differences of donors or patients. Long-term storage of clin. NK cell lots to allow repeated clin. applications remains an addnl. challenge. In contrast, the established and well-characterized cell line NK-92 can be easily and reproducibly expanded from a good manufg. practice (GMP)-compliant cryopreserved master cell bank. Moreover, no cost-intensive cell purifn. methods are required. To date, NK-92 has been intensively studied. The cells displayed superior cytotoxicity against a no. of tumor types tested, which was confirmed in preclin. mouse studies. Subsequent clin. testing demonstrated safety of NK-92 infusions even at high doses. Despite the phase I nature of the trials conducted so far, some efficacy was noted, particularly against lung tumors. Furthermore, to overcome tumor resistance and for specific targeting, NK-92 has been engineered to express a no. of different chimeric antigen receptors (CARs), including targeting, for example, CD19 or CD20 (anti-B cell malignancies), CD38 (anti-myeloma) or human epidermal growth factor receptor 2 (HER2; ErbB2; anti-epithelial cancers). The concept of an NK cell line as an allogeneic cell therapeutic produced 'off-the-shelf' on demand holds great promise for the development of effective treatments.
- 30Obermeyer, A. C.; Jarman, J. B.; Francis, M. B. N-Terminal Modification of Proteins with O-Aminophenols. J. Am. Chem. Soc. 2014, 136, 9572– 9579, DOI: 10.1021/ja500728c[ACS Full Text
], [CAS], Google Scholar
30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtVGlu77L&md5=ab16257753b27283d65e3d787b0bff7fN-Terminal Modification of Proteins with o-AminophenolsObermeyer, Allie C.; Jarman, John B.; Francis, Matthew B.Journal of the American Chemical Society (2014), 136 (27), 9572-9579CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The synthetic modification of proteins plays an important role in chem. biol. and biomaterials science. These fields provide a const. need for chem. tools that can introduce new functionality in specific locations on protein surfaces. In this work, an oxidative strategy is demonstrated for the efficient modification of N-terminal residues on peptides and N-terminal proline residues on proteins. The strategy uses o-aminophenols or o-catechols that are oxidized to active coupling species in situ using potassium ferricyanide. Peptide screening results have revealed that many N-terminal amino acids can participate in this reaction, and that proline residues are particularly reactive. When applied to protein substrates, the reaction shows a stronger requirement for the proline group. Key advantages of the reaction include its fast second-order kinetics and ability to achieve site-selective modification in a single step using low concns. of reagent. Although free cysteines are also modified by the coupling reaction, they can be protected through disulfide formation and then liberated after N-terminal coupling is complete. This allows access to doubly functionalized bioconjugates that can be difficult to access using other methods. - 31Hurben, A. K.; Erber, L. N.; Tretyakova, N. Y.; Doran, T. M. Proteome-Wide Profiling of Cellular Targets Modified by Dopamine Metabolites Using a Bio-Orthogonally Functionalized Catecholamine. ACS Chem. Biol. 2021, 16, 2581– 2594, DOI: 10.1021/acschembio.1c00629[ACS Full Text
], [CAS], Google Scholar
31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitlOhur%252FF&md5=a4b345179f27c8e8b385dbcaffbb3b64Proteome-Wide Profiling of Cellular Targets Modified by Dopamine Metabolites Using a Bio-Orthogonally Functionalized CatecholamineHurben, Alexander K.; Erber, Luke N.; Tretyakova, Natalia Y.; Doran, Todd M.ACS Chemical Biology (2021), 16 (11), 2581-2594CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Selective death of midbrain dopaminergic neurons is a hallmark pathol. of Parkinson's disease (PD), but the mol. mechanisms that initiate the cascade of events resulting in neurodegeneration in PD remain unclear. Compelling evidence suggests that dysregulation of dopamine (DA) induces neuronal stress and damage responses that are operative processes in striatal degeneration preceding PD-like symptoms. Improper DA sequestration to vesicles raises cytosolic DA levels, which is rapidly converted into electrophilic dopaquinone species (DQs) that react readily with protein nucleophiles forming covalent modifications that alter the native structure and function of proteins. These so-called DA-protein adducts (DPAs) have been reported to play a role in neurotoxicity, and their abundance with respect to neurodegeneration has been linked to clin. and pathol. features of PD that suggest that they play a causal role in PD pathogenesis. Therefore, characterizing DPAs is a crit. first step in understanding the susceptibility of midbrain dopaminergic neurons during PD. To help achieve this goal, we report here a novel DA-mimetic (DAyne) contg. a biorthogonal alkyne handle that exhibits a reactivity profile similar to DA in aq. buffers. By linking DPAs formed with DAyne to a fluorescent reporter mol., DPAs were visualized in fixed cells and within lysates. DAyne enabled global mapping of cellular proteins affected by DQ modification and their bioactive pathways through enrichment. Our proteomic profiling of DPAs in neuronal SH-SY5Y cells indicates that proteins susceptible to DPA formation are extant throughout the proteome, potentially influencing several diverse biol. pathways involved in PD such as endoplasmic reticulum (ER) stress, cytoskeletal instability, proteotoxicity, and clathrin function. We validated that a protein involved in the ER stress pathway, protein disulfide isomerase 3 (PDIA3), which was enriched in our chemoproteomic anal., is functionally inhibited by DA, providing evidence that dysregulated cellular DA may induce or exacerbate ER stress. Thus, DAyne provided new mechanistic insights into DA toxicity that may be obsd. during PD by enabling characterization of DPAs generated reproducibly at physiol. relevant quinone exposures. We anticipate our design and application of this reactivity-based probe will be generally applicable for clarifying mechanisms of metabolic quinone toxicity. - 32Bruce, V. J.; Lopez-Islas, M.; McNaughton, B. R. Resurfaced Cell-Penetrating Nanobodies: A Potentially General Scaffold for Intracellularly Targeted Protein Discovery. Protein Sci. 2016, 25, 1129– 1137, DOI: 10.1002/pro.2926[Crossref], [PubMed], [CAS], Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XltlWhsrc%253D&md5=849322d18d9dea14cd01aa3977f7b72bResurfaced cell-penetrating nanobodies: A potentially general scaffold for intracellularly targeted protein discoveryBruce, Virginia J.; Lopez-Islas, Monica; McNaughton, Brian R.Protein Science (2016), 25 (6), 1129-1137CODEN: PRCIEI; ISSN:1469-896X. (Wiley-Blackwell)By virtue of their size, functional group diversity, and complex structure, proteins can often recognize and modulate disease-relevant macromols. that present a challenge to small-mol. reagents. Addnl., high-throughput screening and evolution-based methods often make the discovery of new protein binders simpler than the analogous small-mol. discovery process. However, most proteins do not cross the lipid bilayer membrane of mammalian cells. This largely limits the scope of protein therapeutics and basic research tools to those targeting disease-relevant receptors on the cell surface or extracellular matrix. Previously, researchers have shown that cationic resurfacing of proteins can endow cell penetration. However, in our experience, many proteins are not amenable to such extensive mutagenesis. Here, we report that nanobodies-a small and stable protein that can be evolved to recognize virtually any disease-relevant receptor-are amenable to cationic resurfacing, which results in cell internalization. Once internalized, these nanobodies access the cytosol. Polycationic resurfacing does not appreciably alter the structure, expression, and function (target recognition) of a previously reported GFP-binding nanobody, and multiple nanobody scaffolds are amenable to polycationic resurfacing. Given this, we propose that polycationic resurfaced cell-penetrating nanobodies might represent a general scaffold for intracellularly targeted protein drug discovery.
- 33Lyon, R. Drawing Lessons from the Clinical Development of Antibody-Drug Conjugates. Drug Discovery Today Technologies. 2018, 30, 105– 109, DOI: 10.1016/j.ddtec.2018.10.001[Crossref], [PubMed], [CAS], Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cnisVSlsA%253D%253D&md5=dfdb2af91ce423b8912dc33f9187b35fDrawing lessons from the clinical development of antibody-drug conjugatesLyon RobertDrug discovery today. Technologies (2018), 30 (), 105-109 ISSN:.The antibody-drug conjugate (ADC) field has seen a remarkable expansion in the number of entrants in clinical studies. Many of these agents employ newer conjugation technologies that have been developed over the last decade that confer various attributes to the ADCs prepared with them, including stability, potency, and homogeneity. In many cases, these new ADCs appear demonstrably superior to earlier technologies in preclinical models of activity and toxicology, but the degree to which these improvements will translate to the clinic is only starting to be seen. Many of these technologies are now competing head-to-head by targeting the same antigen in similar patient populations, allowing for a direct comparison of their clinical performance properties. As lessons from these experiences feed back into discovery research, future iterations of ADC design may be expected to bring improved therapeutics into the clinic.
- 34Bauer, S.; Groh, V.; Wu, J.; Steinle, A.; Phillips, J. H.; Lanier, L. L.; Spies, T. Activation of NK Cells and T Cells by NKG2D, a Receptor for Stress-Inducible MICA. Science. 1999, 285, 727– 729, DOI: 10.1126/science.285.5428.727[Crossref], [PubMed], [CAS], Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXltVehsro%253D&md5=296b66b9314dcaaa0bb23d448df2e186Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICABauer, Stefan; Groh, Veronika; Phillips, Joseph H.; Lainer, Lewis L.; Spies, ThomasScience (Washington, D. C.) (1999), 285 (5428), 727-729CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Stress-inducible MICA, a distant homolog of major histocompatibility complex (MHC) class I, functions as an antigen for γδ T cells and is frequently expressed in epithelial tumors. A receptor for MICA was detected on most γδ T cells, CD8+ αβ T cells, and natural killer (NK) cells and was identified as NKG2D. Effector cells from all these subsets could be a stimulated by ligation of NKG2D. Engagement of NKG2D activated cytolytic responses of γδ T cells and NK cells against transfectants and epithelial tumor cells expressing MICA. These results define an activating immunoreceptor-MHC ligand interaction that may promote antitumor NK and T cell responses.
Supporting Information
Supporting Information
ARTICLE SECTIONSThe Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acscentsci.1c01265.
Raw peptide lists for small-molecule proteomics (XLSX)
Unique peptides for small-molecule proteomics (XLSX)
Full experimental details and cloning procedures (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.