1
|
Minoshima M, Reja SI, Hashimoto R, Iijima K, Kikuchi K. Hybrid Small-Molecule/Protein Fluorescent Probes. Chem Rev 2024; 124:6198-6270. [PMID: 38717865 DOI: 10.1021/acs.chemrev.3c00549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Hybrid small-molecule/protein fluorescent probes are powerful tools for visualizing protein localization and function in living cells. These hybrid probes are constructed by diverse site-specific chemical protein labeling approaches through chemical reactions to exogenous peptide/small protein tags, enzymatic post-translational modifications, bioorthogonal reactions for genetically incorporated unnatural amino acids, and ligand-directed chemical reactions. The hybrid small-molecule/protein fluorescent probes are employed for imaging protein trafficking, conformational changes, and bioanalytes surrounding proteins. In addition, fluorescent hybrid probes facilitate visualization of protein dynamics at the single-molecule level and the defined structure with super-resolution imaging. In this review, we discuss development and the bioimaging applications of fluorescent probes based on small-molecule/protein hybrids.
Collapse
Affiliation(s)
- Masafumi Minoshima
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1, Yamadaoka, Suita, Osaka 5650871, Japan
| | - Shahi Imam Reja
- Immunology Frontier Research Center, Osaka University, 2-1, Yamadaoka, Suita, Osaka 5650871, Japan
| | - Ryu Hashimoto
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1, Yamadaoka, Suita, Osaka 5650871, Japan
| | - Kohei Iijima
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1, Yamadaoka, Suita, Osaka 5650871, Japan
| | - Kazuya Kikuchi
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1, Yamadaoka, Suita, Osaka 5650871, Japan
| |
Collapse
|
2
|
Jin C, Zongo AWS, Du H, Lu Y, Yu N, Nie X, Ma A, Ye Q, Xiao H, Meng X. Gardenia ( Gardenia jasminoides Ellis) fruit: a critical review of its functional nutrients, processing methods, health-promoting effects, comprehensive application and future tendencies. Crit Rev Food Sci Nutr 2023:1-28. [PMID: 37882781 DOI: 10.1080/10408398.2023.2270530] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
Gardenia fruit (GF) is the mature fruit of Gardenia jasminoides Ellis, boasting a rich array of nutrients and phytochemicals. Over time, GF has been extensively utilized in both food and medicinal contexts. In recent years, numerous studies have delved into the chemical constituents of GF and their associated pharmacological activities, encompassing its phytochemical composition and health-promoting properties. This review aims to provide a critical and comprehensive summary of GF research, covering nutrient content, extraction technologies, and potential health benefits, offering new avenues for future investigations and highlighting its potential as an innovative food resource. Additionally, the review proposes novel industrial applications for GF, such as utilizing gardenia yellow/red/blue pigments in the food industry and incorporating it with other herbs in traditional Chinese medicine. By addressing current challenges in developing GF-related products, this work provides insights for potential applications in the cosmetics, food, and health products industries. Notably, there is a need for the development of more efficient extraction methods to harness the nutritional components of GF fully. Further research is needed to understand the specific molecular mechanisms underlying its bioactivities. Exploring advanced processing techniques to create innovative GF-derived products will show great promise for the future.
Collapse
Affiliation(s)
- Chengyu Jin
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Abel Wend-Soo Zongo
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Hengjun Du
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Yuanchao Lu
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Ningxiang Yu
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Xiaohua Nie
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Ashton Ma
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
- Phillips Academy Andover, Andover, MA, USA
| | - Qin Ye
- College of Biology and Environmental Engineering, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Xianghe Meng
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Regulatory T Cell Depletion Using a CRISPR Fc-Optimized CD25 Antibody. Int J Mol Sci 2022; 23:ijms23158707. [PMID: 35955841 PMCID: PMC9369266 DOI: 10.3390/ijms23158707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 11/25/2022] Open
Abstract
Regulatory T cells (Tregs) are major drivers behind immunosuppressive mechanisms and present a major hurdle for cancer therapy. Tregs are characterized by a high expression of CD25, which is a potentially valuable target for Treg depletion to alleviate immune suppression. The preclinical anti-CD25 (αCD25) antibody, clone PC-61, has met with modest anti-tumor activity due to its capacity to clear Tregs from the circulation and lymph nodes, but not those that reside in the tumor. The optimization of the Fc domain of this antibody clone has been shown to enhance the intratumoral Treg depletion capacity. Here, we generated a stable cell line that produced optimized recombinant Treg-depleting antibodies. A genome engineering strategy in which CRISPR-Cas9 was combined with homology-directed repair (CRISPR-HDR) was utilized to optimize the Fc domain of the hybridoma PC-61 for effector functions by switching it from its original rat IgG1 to a mouse IgG2a isotype. In a syngeneic tumor mouse model, the resulting αCD25-m2a (mouse IgG2a isotype) antibody mediated the effective depletion of tumor-resident Tregs, leading to a high effector T cell (Teff) to Treg ratio. Moreover, a combination of αCD25-m2a and an αPD-L1 treatment augmented tumor eradication in mice, demonstrating the potential for αCD25 as a cancer immunotherapy.
Collapse
|
4
|
Yu MY, Hua ZY, Liao PR, Zheng H, Jin Y, Peng HS, Cui XM, Huang LQ, Yuan Y. Increasing Expression of PnGAP and PnEXPA4 Provides Insights Into the Enlargement of Panax notoginseng Root Size From Qing Dynasty to Cultivation Era. FRONTIERS IN PLANT SCIENCE 2022; 13:878796. [PMID: 35668802 PMCID: PMC9164015 DOI: 10.3389/fpls.2022.878796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/07/2022] [Indexed: 06/15/2023]
Abstract
Root size is a key trait in plant cultivation and can be influenced by the cultivation environment. However, physical evidence of root size change in a secular context is scarce due to the difficulty in preserving ancient root samples, and how they were modified during the domestication and cultivation stays unclear. About 100 ancient root samples of Panax notoginseng, preserved as tribute in the Palace Museum (A.D. 1636 to 1912, Qing dynasty), provided an opportunity to investigate the root size changes during the last 100 years of cultivation. The dry weight of ancient root samples (~120 tou samples, tou represents number of roots per 500 g dry weight) is 0.22-fold of the modern samples with the biggest size (20 tou samples). Transcriptome analysis revealed that PnGAP and PnEXPA4 were highly expressed in 20 tou samples, compared with the 120 tou samples, which might contribute to the thicker cell wall and a higher content of lignin, cellulose, and callose in 20 tou samples. A relatively lower content of dencichine and higher content of ginsenoside Rb1 in 20 tou samples are also consistent with higher expression of ginsenoside biosynthesis-related genes. PnPHL8 was filtrated through transcriptome analysis, which could specifically bind the promoters of PnGAP, PnCYP716A47, and PnGGPPS3, respectively. The results in this study represent the first physical evidence of root size changes in P. notoginseng in the last 100 years of cultivation and contribute to a comprehensive understanding of how the cultivation environment affected root size, chemical composition, and clinical application.
Collapse
Affiliation(s)
- Mu-Yao Yu
- State Key Laboratory Breeding Base of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhong-Yi Hua
- State Key Laboratory Breeding Base of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Pei-Ran Liao
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Han Zheng
- State Key Laboratory Breeding Base of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yan Jin
- State Key Laboratory Breeding Base of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hua-Sheng Peng
- State Key Laboratory Breeding Base of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiu-Ming Cui
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Lu-Qi Huang
- State Key Laboratory Breeding Base of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuan Yuan
- State Key Laboratory Breeding Base of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
5
|
Abstract
Antibodies, particularly of the immunoglobulin G (IgG) isotype, are a group of biomolecules that are extensively used as affinity reagents for many applications in research, disease diagnostics, and therapy. Most of these applications require antibodies to be modified with specific functional moieties, including fluorophores, drugs, and proteins. Thus, a variety of methodologies have been developed for the covalent labeling of antibodies. The most common methods stably attach functional molecules to lysine or cysteine residues, which unavoidably results in heterogeneous products that cannot be further purified. In an effort to prepare homogeneous antibody conjugates, bioorthogonal handles have been site-specifically introduced via enzymatic treatment, genetic code expansion, or genetically encoded tagging, followed by functionalization using bioorthogonal conjugation reactions. The resulting homogeneous products have proven superior to their heterogeneous counterparts for both in vitro and in vivo usage. Nevertheless, additional chemical treatment or protein engineering of antibodies is required for incorporation of the bioorthogonal handles, processes that often affect antibody folding, stability, and/or production yield and cost. Accordingly, concurrent with advances in the fields of bioorthogonal chemistry and protein engineering, there is growing interest in site-specifically labeling native (nonengineered) antibodies without chemical or enzymatic treatments. In this review, we highlight recent strategies for producing site-specific native antibody conjugates and provide a comprehensive summary of the merits and disadvantages of these strategies.
Collapse
Affiliation(s)
- Kuan-Lin Wu
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Chenfei Yu
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Catherine Lee
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Chao Zuo
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Zachary T Ball
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Han Xiao
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
- Department of Biosciences, Rice University, 6100 Main Street, Houston, Texas 77005, United States
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| |
Collapse
|
6
|
Lu Z, Truex NL, Melo MB, Cheng Y, Li N, Irvine DJ, Pentelute BL. IgG-Engineered Protective Antigen for Cytosolic Delivery of Proteins into Cancer Cells. ACS CENTRAL SCIENCE 2021; 7:365-378. [PMID: 33655074 PMCID: PMC7908032 DOI: 10.1021/acscentsci.0c01670] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Indexed: 05/05/2023]
Abstract
Therapeutic immunotoxins composed of antibodies and bacterial toxins provide potent activity against malignant cells, but joining them with a defined covalent bond while maintaining the desired function is challenging. Here, we develop novel immunotoxins by dovetailing full-length immunoglobulin G (IgG) antibodies and nontoxic anthrax proteins, in which the C terminus of the IgG heavy chain is connected to the side chain of anthrax toxin protective antigen. This strategy enabled efficient conjugation of protective antigen variants to trastuzumab (Tmab) and cetuximab (Cmab) antibodies. The conjugates effectively perform intracellular delivery of edema factor and N terminus of lethal factor (LFN) fused with diphtheria toxin and Ras/Rap1-specific endopeptidase. Each conjugate shows high specificity for cells expressing human epidermal growth factor receptor 2 (HER2) and epidermal growth factor receptor (EGFR), respectively, and potent activity across six Tmab- and Cmab-resistant cell lines. The conjugates also exhibit increased pharmacokinetics and pronounced in vivo safety, which shows promise for further therapeutic development.
Collapse
Affiliation(s)
- Zeyu Lu
- Department
of Chemistry, Massachusetts Institute of
Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Nicholas L. Truex
- Department
of Chemistry, Massachusetts Institute of
Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Mariane B. Melo
- The
Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
- Ragon
Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Yiran Cheng
- Department
of Chemistry, Massachusetts Institute of
Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Na Li
- The
Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| | - Darrell J. Irvine
- The
Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
- Ragon
Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Department
of Materials Science and Engineering, Massachusetts
Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Department
of Biological Engineering, Massachusetts
Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Howard Hughes
Medical Institute, 4000
Jones Bridge Road, Chevy Chase, Maryland 20815, United
States
| | - Bradley L. Pentelute
- Department
of Chemistry, Massachusetts Institute of
Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- The
Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
- Center
for Environmental Health Sciences, Massachusetts
Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Broad
Institute of MIT and Harvard, 415 Main Street, Cambridge, Massachusetts 02142, United States
- E-mail:
| |
Collapse
|
7
|
Zhang B, Vidanapathirana SM, Greineder CF. Site-Specific Modification of Single-Chain Affinity Ligands for Fluorescence Labeling, Radiolabeling, and Bioconjugation. Methods Mol Biol 2021; 2355:163-173. [PMID: 34386959 PMCID: PMC9289842 DOI: 10.1007/978-1-0716-1617-8_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Single-chain protein affinity ligands are recombinant polypeptides that recreate the antigen-binding site of parental, monoclonal antibodies (mAbs) or present unique binding surfaces derived from display technologies, computational design, or other approaches. These diverse ligands have several advantages over full-length mAbs as agents for delivery of small molecule, protein, and nanoparticle cargoes to desired sites in the body. However, they present unique challenges for modification and bioconjugation. Fusion of a LPXTGG motif, or "sortag," and a 5-amino acid, flexible linker to the C-terminus of these affinity ligands enables high-efficiency transpeptidation by the bacterial enzyme, Sortase A, and site-specific addition of fluorophores, radiolabels, or functional groups for oriented and stoichiometrically controlled bioconjugation. We describe in detail this method and address several challenges and pitfalls in the purification and characterization of modified single-chain affinity ligands.
Collapse
Affiliation(s)
- Boya Zhang
- Departments of Pharmacology and Emergency Medicine, University of Michigan, Ann Arbor, MI, USA
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Sachith M Vidanapathirana
- Departments of Pharmacology and Emergency Medicine, University of Michigan, Ann Arbor, MI, USA
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Colin F Greineder
- Departments of Pharmacology and Emergency Medicine, University of Michigan, Ann Arbor, MI, USA.
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
8
|
Sue CK, McConnell SA, Ellis-Guardiola K, Muroski J, McAllister RA, Yu J, Alvarez AI, Chang C, Ogorzalek Loo RR, Loo JA, Ton-That H, Clubb RT. Kinetics and Optimization of the Lysine-Isopeptide Bond Forming Sortase Enzyme from Corynebacterium diphtheriae. Bioconjug Chem 2020; 31:1624-1634. [PMID: 32396336 PMCID: PMC8153732 DOI: 10.1021/acs.bioconjchem.0c00163] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Site-specifically modified protein bioconjugates have important applications in biology, chemistry, and medicine. Functionalizing specific protein side chains with enzymes using mild reaction conditions is of significant interest, but remains challenging. Recently, the lysine-isopeptide bond forming activity of the sortase enzyme that builds surface pili in Corynebacterium diphtheriae (CdSrtA) has been reconstituted in vitro. A mutationally activated form of CdSrtA was shown to be a promising bioconjugating enzyme that can attach Leu-Pro-Leu-Thr-Gly peptide fluorophores to a specific lysine residue within the N-terminal domain of the SpaA protein (NSpaA), enabling the labeling of target proteins that are fused to NSpaA. Here we present a detailed analysis of the CdSrtA catalyzed protein labeling reaction. We show that the first step in catalysis is rate limiting, which is the formation of the CdSrtA-peptide thioacyl intermediate that subsequently reacts with a lysine ε-amine in NSpaA. This intermediate is surprisingly stable, limiting spurious proteolysis of the peptide substrate. We report the discovery of a new enzyme variant (CdSrtAΔ) that has significantly improved transpeptidation activity, because it completely lacks an inhibitory polypeptide appendage ("lid") that normally masks the active site. We show that the presence of the lid primarily impairs formation of the thioacyl intermediate and not the recognition of the NSpaA substrate. Quantitative measurements reveal that CdSrtAΔ generates its cross-linked product with a catalytic turnover number of 1.4 ± 0.004 h-1 and that it has apparent KM values of 0.16 ± 0.04 and 1.6 ± 0.3 mM for its NSpaA and peptide substrates, respectively. CdSrtAΔ is 7-fold more active than previously studied variants, labeling >90% of NSpaA with peptide within 6 h. The results of this study further improve the utility of CdSrtA as a protein labeling tool and provide insight into the enzyme catalyzed reaction that underpins protein labeling and pilus biogenesis.
Collapse
Affiliation(s)
- Christopher K. Sue
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
- UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
| | - Scott A. McConnell
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
- UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
| | - Ken Ellis-Guardiola
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
- UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
| | - John Muroski
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
- UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
| | - Rachel A. McAllister
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
- UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
| | - Justin Yu
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
- UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
| | - Ana I. Alvarez
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
- UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
| | - Chungyu Chang
- Molecular Biology Institute and the University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
- Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
| | - Rachel R. Ogorzalek Loo
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
- UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
| | - Joseph A. Loo
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
- UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
- Molecular Biology Institute and the University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
| | - Hung Ton-That
- Molecular Biology Institute and the University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
- Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
| | - Robert T. Clubb
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
- UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
- Molecular Biology Institute and the University of California, Los Angeles, 611 Charles Young Drive East, Los Angeles, CA 90095
| |
Collapse
|
9
|
Gu H, Ghosh S, Staples RJ, Bane SL. β-Hydroxy-Stabilized Boron-Nitrogen Heterocycles Enable Rapid and Efficient C-Terminal Protein Modification. Bioconjug Chem 2019; 30:2604-2613. [PMID: 31483610 DOI: 10.1021/acs.bioconjchem.9b00534] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Bioorthogonal chemistry has enabled the development of bioconjugates in physiological environments while averting interference from endogenous biomolecules. Reactions between carbonyl-containing molecules and alkoxyamines or hydrazines have experienced a resurgence in popularity in bioorthogonal chemistry owing to advances that allow the reactions to occur under physiological conditions. In particular, ortho-carbonyl-substituted phenylboronic acids (CO-PBAs) exhibit greatly accelerated rates of hydrazone and oxime formation via intramolecular Lewis acid catalysis. Unfortunately, the rate of the reverse reaction is also increased, yielding a kinetically less stable bioconjugate. When the substrate is a hydrazine derivative, an intramolecular reaction between the boronic acid and the hydrazone can lead to the formation of a heterocycle containing a boron-nitrogen bond. We have shown previously that α-amino hydrazides undergo rapid reaction with CO-PBAs to form highly stable, tricyclic products, and that this reaction is orthogonal to the popular azide-alkyne and tetrazine-alkene reactions. In this work, we explore a series of heteroatom-substituted hydrazides for their ability to form tricyclic products with two CO-PBAs, 2-formylphenylboronic acid (2fPBA), and 2-acetylphenylboronic acid (AcPBA). In particular, highly stable products were formed using β-hydroxy hydrazides and 2fPBA. C-Terminal β-hydroxy hydrazide proteins are available using conventional biochemical methods, which alleviates one of the difficulties with applications of bioorthogonal chemical reactions: site-specific incorporation of a reactive group into the biomolecular target. Using sortase-mediated ligation (SML), C-terminal threonine and serine hydrazides were appended to a model eGFP protein in high yield. Subsequent labeling with 2fPBA functionalized probes could be performed quickly and quantitatively at neutral pH using micromolar concentrations of reactants. The SML process was applied directly to an expressed protein in cellular extract, and the C-terminal modified target protein was selectively immobilized using 2fPBA-agarose. Elution from the agarose yielded a highly pure protein that retained the hydrazide functionality. This strategy should be generally applicable for rapid, efficient site-specific protein labeling, protein immobilization, and preparation of highly pure functionalized proteins.
Collapse
Affiliation(s)
- Han Gu
- Department of Chemistry , Binghamton University, State University of New York , Binghamton , New York 13902 , United States
| | - Saptarshi Ghosh
- Department of Chemistry , Binghamton University, State University of New York , Binghamton , New York 13902 , United States
| | - Richard J Staples
- Department of Chemistry and Chemical Biology , Michigan State University , East Lansing , Michigan 48824 , United States
| | - Susan L Bane
- Department of Chemistry , Binghamton University, State University of New York , Binghamton , New York 13902 , United States
| |
Collapse
|
10
|
Liu W, Zhao W, Bai X, Jin S, Li Y, Qiu C, Pan L, Ding D, Xu Y, Zhou Z, Chen S. High antitumor activity of Sortase A-generated anti-CD20 antibody fragment drug conjugates. Eur J Pharm Sci 2019; 134:81-92. [DOI: 10.1016/j.ejps.2019.04.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 03/27/2019] [Accepted: 04/11/2019] [Indexed: 12/29/2022]
|
11
|
Gébleux R, Briendl M, Grawunder U, Beerli RR. Sortase A Enzyme-Mediated Generation of Site-Specifically Conjugated Antibody-Drug Conjugates. Methods Mol Biol 2019; 2012:1-13. [PMID: 31161500 DOI: 10.1007/978-1-4939-9546-2_1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Antibody-drug conjugates (ADCs) are highly potent targeted anticancer therapies. They rely on the linking of a selectively targeting antibody moiety with potent cytotoxic payloads to effect antitumoral activity. In recent years, one focus in the ADC field was to create novel methods for site-specifically conjugating payloads to antibodies. The method presented here is based on the S. aureus sortase A-mediated transpeptidation reaction. This method requires antibodies to be engineered in such a way that they possess the sortase recognition pentapeptide motif LPETG on the C-terminus of the immunoglobulin heavy and/or light chains. In addition, the toxin must contain an oligoglycine motif in order to make it a suitable substrate for sortase A. Here we describe a detailed method to conjugate a pentaglycine-modified toxin to the C-termini of LPETG-tagged antibody heavy and light chains using sortase-mediated antibody conjugation (SMAC-Technology™). Highly homogenous, site-specifically conjugated ADCs with controlled drug to antibody ratio and improved overall properties can be obtained with this method.
Collapse
|
12
|
Arkenberg MR, Moore DM, Lin CC. Dynamic control of hydrogel crosslinking via sortase-mediated reversible transpeptidation. Acta Biomater 2019; 83:83-95. [PMID: 30415064 PMCID: PMC6697659 DOI: 10.1016/j.actbio.2018.11.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/31/2018] [Accepted: 11/05/2018] [Indexed: 02/06/2023]
Abstract
Cell-laden hydrogels whose crosslinking density can be dynamically and reversibly tuned are highly sought-after for studying pathophysiological cellular fate processes, including embryogenesis, fibrosis, and tumorigenesis. Special efforts have focused on controlling network crosslinking in poly(ethylene glycol) (PEG) based hydrogels to evaluate the impact of matrix mechanics on cell proliferation, morphogenesis, and differentiation. In this study, we sought to design dynamic PEG-peptide hydrogels that permit cyclic/reversible stiffening and softening. This was achieved by utilizing reversible enzymatic reactions that afford specificity, biorthogonality, and predictable reaction kinetics. To that end, we prepared PEG-peptide conjugates to enable sortase A (SrtA) induced tunable hydrogel crosslinking independent of macromer contents. Uniquely, these hydrogels can be completely degraded by the same enzymatic reactions and the degradation rate can be tuned from hours to days. We further synthesized SrtA-sensitive peptide linker (i.e., KCLPRTGCK) for crosslinking with 8-arm PEG-norbornene (PEG8NB) via thiol-norbornene photocrosslinking. These hydrogels afford diverse softening paradigms through control of network structures during crosslinking or by adjusting enzymatic parameters during on-demand softening. Importantly, user-controlled hydrogel softening promoted spreading of human mesenchymal stem cells (hMSCs) in 3D. Finally, we designed a bis-cysteine-bearing linear peptide flanked with SrtA substrates at the peptide's N- and C-termini (i.e., NH2-GGGCKGGGKCLPRTG-CONH2) to enable cyclic/reversible hydrogel stiffening/softening. We show that matrix stiffening and softening play a crucial role in growth and chemoresistance in pancreatic cancer cells. These results represent the first dynamic hydrogel platform that affords cyclic gel stiffening/softening based on reversible enzymatic reactions. More importantly, the chemical motifs that affords such reversible crosslinking were built-in on the linear peptide crosslinker without any post-synthesis modification. STATEMENT OF SIGNIFICANCE: Cell-laden 'dynamic' hydrogels are typically designed to enable externally stimulated stiffening or softening of the hydrogel network. However, no enzymatic reaction has been used to reversibly control matrix crosslinking. The application of SrtA-mediated transpeptidation in crosslinking and post-gelation modification of biomimetic hydrogels is innovative because of the specificity of the reaction and reversible tunability of crosslinking kinetics. While SrtA has been previously used to crosslink and fully degrade hydrogels, matrix softening and reversible stiffening of cell-laden hydrogels has not been reported. By designing simple peptide substrates, this unique enzymatic reaction can be employed to form a primary network, to gradually soften hydrogels, or to reversibly stiffen hydrogels. As a result, this dynamic hydrogel platform can be used to answer important matrix-related biological questions that are otherwise difficult to address.
Collapse
Affiliation(s)
- Matthew R Arkenberg
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Dustin M Moore
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Chien-Chi Lin
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| |
Collapse
|
13
|
Wöll S, Bachran C, Schiller S, Schröder M, Conrad L, Scherließ R, Swee LK. Sortagging of liposomes with a murine CD11b-specific VHH increases in vitro and in vivo targeting specificity of myeloid cells. Eur J Pharm Biopharm 2019; 134:190-198. [DOI: 10.1016/j.ejpb.2018.11.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/15/2018] [Accepted: 11/16/2018] [Indexed: 10/27/2022]
|
14
|
Zhang Y, Park KY, Suazo KF, Distefano MD. Recent progress in enzymatic protein labelling techniques and their applications. Chem Soc Rev 2018; 47:9106-9136. [PMID: 30259933 PMCID: PMC6289631 DOI: 10.1039/c8cs00537k] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Protein-based conjugates are valuable constructs for a variety of applications. Conjugation of proteins to fluorophores is commonly used to study their cellular localization and the protein-protein interactions. Modification of therapeutic proteins with either polymers or cytotoxic moieties greatly enhances their pharmacokinetics or potency. To label a protein of interest, conventional direct chemical reaction with the side-chains of native amino acids often yields heterogeneously modified products. This renders their characterization complicated, requires difficult separation steps and may impact protein function. Although modification can also be achieved via the insertion of unnatural amino acids bearing bioorthogonal functional groups, these methods can have lower protein expression yields, limiting large scale production. As a site-specific modification method, enzymatic protein labelling is highly efficient and robust under mild reaction conditions. Significant progress has been made over the last five years in modifying proteins using enzymatic methods for numerous applications, including the creation of clinically relevant conjugates with polymers, cytotoxins or imaging agents, fluorescent or affinity probes to study complex protein interaction networks, and protein-linked materials for biosensing. This review summarizes developments in enzymatic protein labelling over the last five years for a panel of ten enzymes, including sortase A, subtiligase, microbial transglutaminase, farnesyltransferase, N-myristoyltransferase, phosphopantetheinyl transferases, tubulin tyrosin ligase, lipoic acid ligase, biotin ligase and formylglycine generating enzyme.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | | | | | |
Collapse
|
15
|
McConnell SA, Amer BR, Muroski J, Fu J, Chang C, Ogorzalek Loo RR, Loo JA, Osipiuk J, Ton-That H, Clubb RT. Protein Labeling via a Specific Lysine-Isopeptide Bond Using the Pilin Polymerizing Sortase from Corynebacterium diphtheriae. J Am Chem Soc 2018; 140:8420-8423. [PMID: 29927249 PMCID: PMC6230430 DOI: 10.1021/jacs.8b05200] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Proteins that are site-specifically modified with peptides and chemicals can be used as novel therapeutics, imaging tools, diagnostic reagents and materials. However, there are few enzyme-catalyzed methods currently available to selectively conjugate peptides to internal sites within proteins. Here we show that a pilus-specific sortase enzyme from Corynebacterium diphtheriae (CdSrtA) can be used to attach a peptide to a protein via a specific lysine-isopeptide bond. Using rational mutagenesis we created CdSrtA3M, a highly activated cysteine transpeptidase that catalyzes in vitro isopeptide bond formation. CdSrtA3M mediates bioconjugation to a specific lysine residue within a fused domain derived from the corynebacterial SpaA protein. Peptide modification yields greater than >95% can be achieved. We demonstrate that CdSrtA3M can be used in concert with the Staphylococcus aureus SrtA enzyme, enabling dual, orthogonal protein labeling via lysine-isopeptide and backbone-peptide bonds.
Collapse
Affiliation(s)
- Scott A. McConnell
- Department of Chemistry and Biochemistry, UCLA-DOE Institute for Genomics and Proteomics and the Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Brendan R. Amer
- Department of Chemistry and Biochemistry, UCLA-DOE Institute for Genomics and Proteomics and the Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - John Muroski
- Department of Chemistry and Biochemistry, UCLA-DOE Institute for Genomics and Proteomics and the Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Janine Fu
- Department of Chemistry and Biochemistry, UCLA-DOE Institute for Genomics and Proteomics and the Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Chungyu Chang
- Department of Microbiology & Molecular Genetics, University of Texas Health Science Center, Houston, TX, USA
| | - Rachel R. Ogorzalek Loo
- Department of Chemistry and Biochemistry, UCLA-DOE Institute for Genomics and Proteomics and the Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Joseph A. Loo
- Department of Chemistry and Biochemistry, UCLA-DOE Institute for Genomics and Proteomics and the Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Jerzy Osipiuk
- Structural Biology Center, Argonne National Laboratory, Argonne, IL, USA
| | - Hung Ton-That
- Department of Microbiology & Molecular Genetics, University of Texas Health Science Center, Houston, TX, USA
| | - Robert T. Clubb
- Department of Chemistry and Biochemistry, UCLA-DOE Institute for Genomics and Proteomics and the Molecular Biology Institute, University of California, Los Angeles, CA, USA
| |
Collapse
|
16
|
Liu Y, Wu HC, Bhokisham N, Li J, Hong KL, Quan DN, Tsao CY, Bentley WE, Payne GF. Biofabricating Functional Soft Matter Using Protein Engineering to Enable Enzymatic Assembly. Bioconjug Chem 2018; 29:1809-1822. [PMID: 29745651 PMCID: PMC7045599 DOI: 10.1021/acs.bioconjchem.8b00197] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Biology often provides the inspiration for functional soft matter, but biology can do more: it can provide the raw materials and mechanisms for hierarchical assembly. Biology uses polymers to perform various functions, and biologically derived polymers can serve as sustainable, self-assembling, and high-performance materials platforms for life-science applications. Biology employs enzymes for site-specific reactions that are used to both disassemble and assemble biopolymers both to and from component parts. By exploiting protein engineering methodologies, proteins can be modified to make them more susceptible to biology's native enzymatic activities. They can be engineered with fusion tags that provide (short sequences of amino acids at the C- and/or N- termini) that provide the accessible residues for the assembling enzymes to recognize and react with. This "biobased" fabrication not only allows biology's nanoscale components (i.e., proteins) to be engineered, but also provides the means to organize these components into the hierarchical structures that are prevalent in life.
Collapse
Affiliation(s)
| | - Hsuan-Chen Wu
- Department of Biochemical Science and Technology , National Taiwan University , Taipei City , Taiwan
| | | | | | - Kai-Lin Hong
- Department of Biochemical Science and Technology , National Taiwan University , Taipei City , Taiwan
| | | | | | | | | |
Collapse
|
17
|
Braun AC, Gutmann M, Lühmann T, Meinel L. Bioorthogonal strategies for site-directed decoration of biomaterials with therapeutic proteins. J Control Release 2018; 273:68-85. [PMID: 29360478 DOI: 10.1016/j.jconrel.2018.01.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/16/2018] [Accepted: 01/17/2018] [Indexed: 01/04/2023]
Abstract
Emerging strategies targeting site-specific protein modifications allow for unprecedented selectivity, fast kinetics and mild reaction conditions with high yield. These advances open exciting novel possibilities for the effective bioorthogonal decoration of biomaterials with therapeutic proteins. Site-specificity is particularly important to the therapeutics' end and translated by targeting specific functional groups or introducing new functional groups into the therapeutic at predefined positions. Biomimetic strategies are designed for modification of therapeutics emulating enzymatic strategies found in Nature. These strategies are suitable for a diverse range of applications - not only for protein-polymer conjugation, particle decoration and surface immobilization, but also for the decoration of complex biomaterials and the synthesis of bioresponsive drug delivery systems. This article reviews latest chemical and enzymatic strategies for the biorthogonal decoration of biomaterials with therapeutic proteins and inter-positioned linker structures. Finally, the numerous reports at the interface of biomaterials, linkers, and therapeutic protein decoration are integrated into practical advice for design considerations intended to support the selection of productive ligation strategies.
Collapse
Affiliation(s)
- Alexandra C Braun
- Institute for Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, DE-97074 Würzburg, Germany
| | - Marcus Gutmann
- Institute for Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, DE-97074 Würzburg, Germany
| | - Tessa Lühmann
- Institute for Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, DE-97074 Würzburg, Germany
| | - Lorenz Meinel
- Institute for Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, DE-97074 Würzburg, Germany.
| |
Collapse
|
18
|
Enzyme-Based Labeling Strategies for Antibody-Drug Conjugates and Antibody Mimetics. Antibodies (Basel) 2018; 7:antib7010004. [PMID: 31544857 PMCID: PMC6698867 DOI: 10.3390/antib7010004] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 01/25/2023] Open
Abstract
Strategies for site-specific modification of proteins have increased in number, complexity, and specificity over the last years. Such modifications hold the promise to broaden the use of existing biopharmaceuticals or to tailor novel proteins for therapeutic or diagnostic applications. The recent quest for next-generation antibody–drug conjugates (ADCs) sparked research into techniques with site selectivity. While purely chemical approaches often impede control of dosage or locus of derivatization, naturally occurring enzymes and proteins bear the ability of co- or post-translational protein modifications at particular residues, thus enabling unique coupling reactions or protein fusions. This review provides a general overview and focuses on chemo-enzymatic methods including enzymes such as formylglycine-generating enzyme, sortase, and transglutaminase. Applications for the conjugation of antibodies and antibody mimetics are reported.
Collapse
|
19
|
Greineder CF, Villa CH, Walsh LR, Kiseleva RY, Hood ED, Khoshnejad M, Warden-Rothman R, Tsourkas A, Muzykantov VR. Site-Specific Modification of Single-Chain Antibody Fragments for Bioconjugation and Vascular Immunotargeting. Bioconjug Chem 2017; 29:56-66. [PMID: 29200285 DOI: 10.1021/acs.bioconjchem.7b00592] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The conjugation of antibodies to drugs and drug carriers improves delivery to target tissues. Widespread implementation and effective translation of this pharmacologic strategy awaits the development of affinity ligands capable of a defined degree of modification and highly efficient bioconjugation without loss of affinity. To date, such ligands are lacking for the targeting of therapeutics to vascular endothelial cells. To enable site-specific, click-chemistry conjugation to therapeutic cargo, we used the bacterial transpeptidase, sortase A, to attach short azidolysine containing peptides to three endothelial-specific single chain antibody fragments (scFv). While direct fusion of a recognition motif (sortag) to the scFv C-terminus generally resulted in low levels of sortase-mediated modification, improved reaction efficiency was observed for one protein, in which two amino acids had been introduced during cloning. This prompted insertion of a short, semi-rigid linker between scFv and sortag. The linker significantly enhanced modification of all three proteins, to the extent that unmodified scFv could no longer be detected. As proof of principle, purified, azide-modified scFv was conjugated to the antioxidant enzyme, catalase, resulting in robust endothelial targeting of functional cargo in vitro and in vivo.
Collapse
Affiliation(s)
- Colin F Greineder
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine and ‡Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania 19104, United States
| | - Carlos H Villa
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine and ‡Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania 19104, United States
| | - Landis R Walsh
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine and ‡Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania 19104, United States
| | - Raisa Y Kiseleva
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine and ‡Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania 19104, United States
| | - Elizabeth D Hood
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine and ‡Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania 19104, United States
| | - Makan Khoshnejad
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine and ‡Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania 19104, United States
| | - Robert Warden-Rothman
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine and ‡Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania 19104, United States
| | - Andrew Tsourkas
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine and ‡Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania 19104, United States
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine and ‡Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
20
|
White DR, Khedri Z, Kiptoo P, Siahaan TJ, Tolbert TJ. Synthesis of a Bifunctional Peptide Inhibitor-IgG1 Fc Fusion That Suppresses Experimental Autoimmune Encephalomyelitis. Bioconjug Chem 2017; 28:1867-1877. [PMID: 28581731 PMCID: PMC5659714 DOI: 10.1021/acs.bioconjchem.7b00175] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Multiple sclerosis (MS) is a neurodegenerative disease that is estimated to affect over 2.3 million people worldwide. The exact cause for this disease is unknown but involves immune system attack and destruction of the myelin protein surrounding the neurons in the central nervous system. One promising class of compounds that selectively prevent the activation of immune cells involved in the pathway leading to myelin destruction are bifunctional peptide inhibitors (BPIs). Treatment with BPIs reduces neurodegenerative symptoms in experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. In this work, as an effort to further improve the bioactivity of BPIs, BPI peptides were conjugated to the N- and C-termini of the fragment crystallizable (Fc) region of the human IgG1 antibody. Initially, the two peptides were conjugated to IgG1 Fc using recombinant DNA technology. However, expression in yeast resulted in low yields and one of the peptides being heavily proteolyzed. To circumvent this problem, the poorly expressed peptide was instead produced by solid phase peptide synthesis and conjugated enzymatically using a sortase-mediated ligation. The sortase-mediated method showed near-complete conjugation yield as observed by SDS-PAGE and mass spectrometry in small-scale reactions. This method was scaled up to obtain sufficient quantities for testing the BPI-Fc fusion in mice induced with EAE. Compared to the PBS-treated control, mice treated with the BPI-Fc fusion showed significantly reduced disease symptoms, did not experience weight loss, and showed reduced de-myelination. These results demonstrate that the BPI peptides were highly active at suppressing EAE when conjugated to the large Fc scaffold in this manner.
Collapse
Affiliation(s)
- Derek R. White
- The Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| | - Zahra Khedri
- The Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
- Ajinomoto Althea Inc., San Diego, California 92121, United States
| | - Paul Kiptoo
- The Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
- Sekisui XenoTech, LLC, Kansas City, Kansas 66103, United States
| | - Teruna J. Siahaan
- The Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| | - Thomas J. Tolbert
- The Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| |
Collapse
|
21
|
Milczek EM. Commercial Applications for Enzyme-Mediated Protein Conjugation: New Developments in Enzymatic Processes to Deliver Functionalized Proteins on the Commercial Scale. Chem Rev 2017. [DOI: 10.1021/acs.chemrev.6b00832] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
22
|
Silvius JR, Leventis R. A Novel “Prebinding” Strategy Dramatically Enhances Sortase-Mediated Coupling of Proteins to Liposomes. Bioconjug Chem 2017; 28:1271-1282. [DOI: 10.1021/acs.bioconjchem.7b00087] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- John R. Silvius
- Department of Biochemistry, McGill University, 3655 Promenade Sir-William-Osler, Montréal, QC, Canada H3G 1A9
| | - Rania Leventis
- Department of Biochemistry, McGill University, 3655 Promenade Sir-William-Osler, Montréal, QC, Canada H3G 1A9
| |
Collapse
|
23
|
Pan L, Zhao W, Lai J, Ding D, Zhang Q, Yang X, Huang M, Jin S, Xu Y, Zeng S, Chou JJ, Chen S. Sortase A-Generated Highly Potent Anti-CD20-MMAE Conjugates for Efficient Elimination of B-Lineage Lymphomas. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:1602267. [PMID: 27873460 DOI: 10.1002/smll.201602267] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 10/18/2016] [Indexed: 06/06/2023]
Abstract
Antibody-drug conjugate (ADC) targeting antigens expressed on the surface of tumor cells are an effective approach for delivering drugs into the cells via antigen-mediated endocytosis. One of the well-known tumor antigens, the CD20 of B-lymphocyte, has long been suggested to be noninternalizing epitope, and is thus not considered a desirable target for ADCs. Here, sortase A (srtA)-mediated transpeptidation is used to specifically conjugate triple glycine-modified monomethyl auristatin E (MMAE), a highly toxic antimitotic agent, to anti-CD20 ofatumumab (OFA) equipped with a short C-terminal LPETG (5 amino acids) tag at heavy chain (HL), which generates ADCs that show extremely strong potency in killing CD20 positive cancer cells. One of the srtA-generated ADCs with a cleavable dipeptide linker (valine-citrulline, vc), OFA-HL-vcMMAE, shows IC50 values ranging from 5 pg mL-1 to 4.1 ng mL-1 against CD20+ lymphoma cells. Confocal laser scanning microscopy confirms that OFA-HL-vcMMAE internalization by Ramos cells is significantly improved compared to OFA alone, consistent with the high antitumor activity of the new ADC. OFA-HL-vcMMAE, at 5 mg kg-1 dose, is able to eliminate tumors with mean volume ≈400 mm3 while no obvious drug-related toxicity is observed. The results show that srtA-generated OFA-MMAE conjugate system provides a viable strategy for targeting CD20+ B lineage lymphomas.
Collapse
Affiliation(s)
- Liqiang Pan
- Institute of Drug Metabolism and Drug Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Wenbin Zhao
- Institute of Drug Metabolism and Drug Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jun Lai
- Institute of Drug Metabolism and Drug Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ding Ding
- HisunPharma (Hangzhou) Co., Ltd, Xialian Village, Xukou Town, Fuyang, Hangzhou, 311404, China
| | - Qian Zhang
- Institute of Drug Metabolism and Drug Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiaoyue Yang
- Institute of Drug Metabolism and Drug Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Minmin Huang
- Institute of Drug Metabolism and Drug Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shijie Jin
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yingchun Xu
- Institute of Drug Metabolism and Drug Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Su Zeng
- Institute of Drug Metabolism and Drug Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - James J Chou
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Shuqing Chen
- Institute of Drug Metabolism and Drug Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
24
|
Enzyme-Based Strategies to Generate Site-Specifically Conjugated Antibody Drug Conjugates. NEXT GENERATION ANTIBODY DRUG CONJUGATES (ADCS) AND IMMUNOTOXINS 2017. [DOI: 10.1007/978-3-319-46877-8_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
25
|
Adumeau P, Sharma SK, Brent C, Zeglis BM. Site-Specifically Labeled Immunoconjugates for Molecular Imaging--Part 2: Peptide Tags and Unnatural Amino Acids. Mol Imaging Biol 2016; 18:153-65. [PMID: 26754791 DOI: 10.1007/s11307-015-0920-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Molecular imaging using radioisotope- or fluorophore-labeled antibodies is increasingly becoming a critical component of modern precision medicine. Yet despite this promise, the vast majority of these immunoconjugates are synthesized via the random coupling of amine-reactive bifunctional probes to lysines within the antibody, a process that can result in heterogeneous and poorly defined constructs with suboptimal pharmacological properties. In an effort to circumvent these issues, the last 5 years have played witness to a great deal of research focused on the creation of effective strategies for the site-specific attachment of payloads to antibodies. These chemoselective modification methods yield immunoconjugates that are more homogenous and better defined than constructs created using traditional synthetic approaches. Moreover, site-specifically labeled immunoconjugates have also been shown to exhibit superior in vivo behavior compared to their randomly modified cousins. The over-arching goal of this two-part review is to provide a broad yet detailed account of the various site-specific bioconjugation approaches that have been used to create immunoconjugates for positron emission tomography (PET), single photon emission computed tomography (SPECT), and fluorescence imaging. In Part 1, we covered site-specific bioconjugation techniques based on the modification of cysteine residues and the chemoenzymatic manipulation of glycans. In Part 2, we will detail two families of bioconjugation approaches that leverage biochemical tools to achieve site-specificity. First, we will discuss modification methods that employ peptide tags either as sites for enzyme-catalyzed ligations or as radiometal coordination architectures. And second, we will examine bioconjugation strategies predicated on the incorporation of unnatural or non-canonical amino acids into antibodies via genetic engineering. Finally, we will compare the advantages and disadvantages of the modification strategies covered in both parts of the review and offer a brief discussion of the overall direction of the field.
Collapse
Affiliation(s)
- Pierre Adumeau
- Department of Chemistry and Biochemistry, Hunter College and the Graduate Center of the City University of New York, 413 East 69th Street, New York, NY, 10021, USA
| | - Sai Kiran Sharma
- Department of Chemistry and Biochemistry, Hunter College and the Graduate Center of the City University of New York, 413 East 69th Street, New York, NY, 10021, USA
| | - Colleen Brent
- Department of Chemistry and Biochemistry, Hunter College and the Graduate Center of the City University of New York, 413 East 69th Street, New York, NY, 10021, USA
| | - Brian M Zeglis
- Department of Chemistry and Biochemistry, Hunter College and the Graduate Center of the City University of New York, 413 East 69th Street, New York, NY, 10021, USA. .,Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
26
|
Domeradzka NE, Werten MWT, Wolf FAD, de Vries R. Protein cross-linking tools for the construction of nanomaterials. Curr Opin Biotechnol 2016; 39:61-67. [DOI: 10.1016/j.copbio.2016.01.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 01/06/2016] [Accepted: 01/11/2016] [Indexed: 12/26/2022]
|
27
|
Rosen CB, Kwant RL, MacDonald JI, Rao M, Francis MB. Capture and Recycling of Sortase A through Site-Specific Labeling with Lithocholic Acid. Angew Chem Int Ed Engl 2016; 55:8585-9. [PMID: 27239057 DOI: 10.1002/anie.201602353] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/26/2016] [Indexed: 11/05/2022]
Abstract
Enzyme-mediated protein modification often requires large amounts of biocatalyst, adding significant costs to the process and limiting industrial applications. Herein, we demonstrate a scalable and straightforward strategy for the efficient capture and recycling of enzymes using a small-molecule affinity tag. A proline variant of an evolved sortase A (SrtA 7M) was N-terminally labeled with lithocholic acid (LA)-an inexpensive bile acid that exhibits strong binding to β-cyclodextrin (βCD). Capture and recycling of the LA-Pro-SrtA 7M conjugate was achieved using βCD-modified sepharose resin. The LA-Pro-SrtA 7M conjugate retained full enzymatic activity, even after multiple rounds of recycling.
Collapse
Affiliation(s)
- Christian B Rosen
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720-1460, USA.,Materials Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Richard L Kwant
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720-1460, USA.,Materials Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - James I MacDonald
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720-1460, USA.,Materials Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Meera Rao
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720-1460, USA.,Materials Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Matthew B Francis
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720-1460, USA. .,Materials Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA.
| |
Collapse
|
28
|
Rosen CB, Kwant RL, MacDonald JI, Rao M, Francis MB. Capture and Recycling of Sortase A through Site‐Specific Labeling with Lithocholic Acid. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201602353] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Christian B. Rosen
- Department of Chemistry University of California, Berkeley Berkeley CA 94720-1460 USA
- Materials Sciences Division Lawrence Berkeley National Laboratory Berkeley CA 94720 USA
| | - Richard L. Kwant
- Department of Chemistry University of California, Berkeley Berkeley CA 94720-1460 USA
- Materials Sciences Division Lawrence Berkeley National Laboratory Berkeley CA 94720 USA
| | - James I. MacDonald
- Department of Chemistry University of California, Berkeley Berkeley CA 94720-1460 USA
- Materials Sciences Division Lawrence Berkeley National Laboratory Berkeley CA 94720 USA
| | - Meera Rao
- Department of Chemistry University of California, Berkeley Berkeley CA 94720-1460 USA
- Materials Sciences Division Lawrence Berkeley National Laboratory Berkeley CA 94720 USA
| | - Matthew B. Francis
- Department of Chemistry University of California, Berkeley Berkeley CA 94720-1460 USA
- Materials Sciences Division Lawrence Berkeley National Laboratory Berkeley CA 94720 USA
| |
Collapse
|
29
|
Massa S, Xavier C, Muyldermans S, Devoogdt N. Emerging site-specific bioconjugation strategies for radioimmunotracer development. Expert Opin Drug Deliv 2016; 13:1149-63. [DOI: 10.1080/17425247.2016.1178235] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Sam Massa
- In vivo Cellular and Molecular Imaging laboratory, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Catarina Xavier
- In vivo Cellular and Molecular Imaging laboratory, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Nick Devoogdt
- In vivo Cellular and Molecular Imaging laboratory, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
30
|
Schumacher D, Hackenberger CPR, Leonhardt H, Helma J. Current Status: Site-Specific Antibody Drug Conjugates. J Clin Immunol 2016; 36 Suppl 1:100-7. [PMID: 27003914 PMCID: PMC4891387 DOI: 10.1007/s10875-016-0265-6] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 03/07/2016] [Indexed: 12/04/2022]
Abstract
Antibody drug conjugates (ADCs), a promising class of cancer biopharmaceuticals, combine the specificity of therapeutic antibodies with the pharmacological potency of chemical, cytotoxic drugs. Ever since the first ADCs on the market, a plethora of novel ADC technologies has emerged, covering as diverse aspects as antibody engineering, chemical linker optimization and novel conjugation strategies, together aiming at constantly widening the therapeutic window for ADCs. This review primarily focuses on novel chemical and biotechnological strategies for the site-directed attachment of drugs that are currently validated for 2nd generation ADCs to promote conjugate homogeneity and overall stability.
Collapse
Affiliation(s)
- Dominik Schumacher
- Chemical Biology and Department of Chemistry, Leibniz-Institut für Molekulare Pharmakologie and Humboldt Universität zu Berlin, Berlin, Germany
| | - Christian P R Hackenberger
- Chemical Biology and Department of Chemistry, Leibniz-Institut für Molekulare Pharmakologie and Humboldt Universität zu Berlin, Berlin, Germany
| | - Heinrich Leonhardt
- Department of Biology II, Ludwig-Maximilians-Universität München and Center for Integrated Protein Science Munich, Planegg-Martinsried, Germany
| | - Jonas Helma
- Department of Biology II, Ludwig-Maximilians-Universität München and Center for Integrated Protein Science Munich, Planegg-Martinsried, Germany.
| |
Collapse
|
31
|
Ismail NF, Lim TS. Site-specific scFv labelling with invertase via Sortase A mechanism as a platform for antibody-antigen detection using the personal glucose meter. Sci Rep 2016; 6:19338. [PMID: 26782912 PMCID: PMC4726117 DOI: 10.1038/srep19338] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 11/17/2015] [Indexed: 12/17/2022] Open
Abstract
Antibody labelling to reporter molecules is gaining popularity due to its many potential applications for diagnostics and therapeutics. However, non-directional bioconjugation methods which are commonly used often results in the loss of target binding capabilities. Therefore, a site-specific enzymatic based bioconjugation such as sortase-mediated transpeptidation allows for a more rapid and efficient method of antibody conjugation for diagnostic applications. Here we describe the utilization of sortase A bioconjugation to conjugate a single chain fragment variable (scFv) to the extracellular invertase (invB) from Zymomonas mobilis with the aim of developing an invertase based immunoassay. In addition, conjugation to enhanced green fluorescent protein (eGFP) was also validated to show the flexibility of the method. The invertase conjugated complex was successfully applied for the detection of antibody-antigen interaction using a personal glucose meter (PGM) for assay readout. The setup was used in both a direct and competitive assay highlighting the robustness of the conjugate for assay development. The method provides an alternative conjugation process to allow easy exchange of antibodies to facilitate rapid development of diagnostic assays for various diseases on the PGM platform.
Collapse
Affiliation(s)
- Nur Faezee Ismail
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Theam Soon Lim
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia
| |
Collapse
|
32
|
Alt K, Paterson BM, Westein E, Rudd SE, Poniger SS, Jagdale S, Ardipradja K, Connell TU, Krippner GY, Nair AKN, Wang X, Tochon-Danguy HJ, Donnelly PS, Peter K, Hagemeyer CE. A versatile approach for the site-specific modification of recombinant antibodies using a combination of enzyme-mediated bioconjugation and click chemistry. Angew Chem Int Ed Engl 2015; 54:7515-9. [PMID: 25962581 DOI: 10.1002/anie.201411507] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 02/24/2015] [Indexed: 12/16/2022]
Abstract
A unique two-step modular system for site-specific antibody modification and conjugation is reported. The first step of this approach uses enzymatic bioconjugation with the transpeptidase Sortase A for incorporation of strained cyclooctyne functional groups. The second step of this modular approach involves the azide-alkyne cycloaddition click reaction. The versatility of the two-step approach has been exemplified by the selective incorporation of fluorescent dyes and a positron-emitting copper-64 radiotracer for fluorescence and positron-emission tomography imaging of activated platelets, platelet aggregates, and thrombi, respectively. This flexible and versatile approach could be readily adapted to incorporate a large array of tailor-made functional groups using reliable click chemistry whilst preserving the activity of the antibody or other sensitive biological macromolecules.
Collapse
Affiliation(s)
- Karen Alt
- Vascular Biotechnology, Baker IDI, Melbourne (Australia).
| | - Brett M Paterson
- School of Chemistry/Bio21 Institute, University of Melbourne (Australia)
| | - Erik Westein
- Atherothrombosis and Vascular Biology, Baker IDI, Melbourne (Australia)
| | - Stacey E Rudd
- School of Chemistry/Bio21 Institute, University of Melbourne (Australia)
| | - Stan S Poniger
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne (Australia)
| | - Shweta Jagdale
- Vascular Biotechnology, Baker IDI, Melbourne (Australia)
| | | | - Timothy U Connell
- School of Chemistry/Bio21 Institute, University of Melbourne (Australia)
| | - Guy Y Krippner
- Vascular Biotechnology, Baker IDI, Melbourne (Australia)
| | - Ashish K N Nair
- Atherothrombosis and Vascular Biology, Baker IDI, Melbourne (Australia)
| | - Xiaowei Wang
- Atherothrombosis and Vascular Biology, Baker IDI, Melbourne (Australia)
| | | | - Paul S Donnelly
- School of Chemistry/Bio21 Institute, University of Melbourne (Australia).
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker IDI, Melbourne (Australia)
| | | |
Collapse
|
33
|
Alt K, Paterson BM, Westein E, Rudd SE, Poniger SS, Jagdale S, Ardipradja K, Connell TU, Krippner GY, Nair AKN, Wang X, Tochon-Danguy HJ, Donnelly PS, Peter K, Hagemeyer CE. A Versatile Approach for the Site-Specific Modification of Recombinant Antibodies Using a Combination of Enzyme-Mediated Bioconjugation and Click Chemistry. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201411507] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
34
|
Bradshaw WJ, Davies AH, Chambers CJ, Roberts AK, Shone CC, Acharya KR. Molecular features of the sortase enzyme family. FEBS J 2015; 282:2097-114. [PMID: 25845800 DOI: 10.1111/febs.13288] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 03/13/2015] [Accepted: 03/28/2015] [Indexed: 01/31/2023]
Abstract
Bacteria possess complex and varying cell walls with many surface exposed proteins. Sortases are responsible for the covalent attachment of specific proteins to the peptidoglycan of the cell wall of Gram-positive bacteria. Sortase A of Staphylococcus aureus, which is seen as the archetypal sortase, has been shown to be essential for pathogenesis and has therefore received much attention as a potential target for novel therapeutics. Being widely present in Gram-positive bacteria, it is likely that other Gram-positive pathogens also require sortases for their pathogenesis. Sortases have also been shown to be of significant use in a range of industrial applications. We review current knowledge of the sortase family in terms of their structures, functions and mechanisms and summarize work towards their use as antibacterial targets and microbiological tools.
Collapse
Affiliation(s)
- William J Bradshaw
- Department of Biology and Biochemistry, University of Bath, UK.,Public Health England, Porton Down, Salisbury, UK
| | | | - Christopher J Chambers
- Department of Biology and Biochemistry, University of Bath, UK.,Public Health England, Porton Down, Salisbury, UK
| | | | | | - K Ravi Acharya
- Department of Biology and Biochemistry, University of Bath, UK
| |
Collapse
|
35
|
Agarwal P, Bertozzi CR. Site-specific antibody-drug conjugates: the nexus of bioorthogonal chemistry, protein engineering, and drug development. Bioconjug Chem 2015; 26:176-92. [PMID: 25494884 PMCID: PMC4335810 DOI: 10.1021/bc5004982] [Citation(s) in RCA: 450] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
![]()
Antibody–drug
conjugates (ADCs) combine the specificity
of antibodies with the potency of small molecules to create targeted
drugs. Despite the simplicity of this concept, generation of clinically
successful ADCs has been very difficult. Over the past several decades,
scientists have learned a great deal about the constraints on antibodies,
linkers, and drugs as they relate to successful construction of ADCs.
Once these components are in hand, most ADCs are prepared by nonspecific
modification of antibody lysine or cysteine residues with drug-linker
reagents, which results in heterogeneous product mixtures that cannot
be further purified. With advances in the fields of bioorthogonal
chemistry and protein engineering, there is growing interest in producing
ADCs by site-specific conjugation to the antibody, yielding more homogeneous
products that have demonstrated benefits over their heterogeneous
counterparts in vivo. Here, we chronicle the development
of a multitude of site-specific conjugation strategies for assembly
of ADCs and provide a comprehensive account of key advances and their
roots in the fields of bioorthogonal chemistry and protein engineering.
Collapse
Affiliation(s)
- Paresh Agarwal
- Departments of Chemistry and ‡Molecular and Cell Biology and §Howard Hughes Medical Institute, University of California , Berkeley, California 94720, United States
| | | |
Collapse
|
36
|
Rashidian M, Dozier JK, Distefano MD. Enzymatic labeling of proteins: techniques and approaches. Bioconjug Chem 2014; 24:1277-94. [PMID: 23837885 DOI: 10.1021/bc400102w] [Citation(s) in RCA: 200] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Site-specific modification of proteins is a major challenge in modern chemical biology due to the large number of reactive functional groups typically present in polypeptides. Because of its importance in biology and medicine, the development of methods for site-specific modification of proteins is an area of intense research. Selective protein modification procedures have been useful for oriented protein immobilization, for studies of naturally occurring post-translational modifications, for creating antibody–drug conjugates, for the introduction of fluorophores and other small molecules on to proteins, for examining protein structure, folding, dynamics, and protein–protein interactions, and for the preparation of protein–polymer conjugates. One of the most important approaches for protein labeling is to incorporate bioorthogonal functionalities into proteins at specific sites via enzymatic reactions. The incorporated tags then enable reactions that are chemoselective, whose functional groups not only are inert in biological media, but also do not occur natively in proteins or other macromolecules. This review article summarizes the enzymatic strategies, which enable site-specific functionalization of proteins with a variety of different functional groups. The enzymes covered in this review include formylglycine generating enzyme, sialyltransferases, phosphopantetheinyltransferases, O-GlcNAc post-translational modification, sortagging, transglutaminase, farnesyltransferase, biotin ligase, lipoic acid ligase, and N-myristoyltransferase.
Collapse
|
37
|
Kline T, Steiner AR, Penta K, Sato AK, Hallam TJ, Yin G. Methods to Make Homogenous Antibody Drug Conjugates. Pharm Res 2014; 32:3480-93. [PMID: 25511917 PMCID: PMC4596908 DOI: 10.1007/s11095-014-1596-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 12/03/2014] [Indexed: 02/06/2023]
Abstract
Antibody drug conjugates (ADCs) have progressed from hypothesis to approved therapeutics in less than 30 years, and the technologies available to modify both the antibodies and the cytotoxic drugs are expanding rapidly. For reasons well reviewed previously, the field is trending strongly toward homogeneous, defined antibody conjugation. In this review we present the antibody and small molecule chemistries that are currently used and being explored to develop specific, homogenous ADCs.
Collapse
Affiliation(s)
- Toni Kline
- Sutro Biopharma, Inc, 310 Utah Ave Ste 150, South San Francisco, California, 94080, USA
| | - Alexander R Steiner
- Sutro Biopharma, Inc, 310 Utah Ave Ste 150, South San Francisco, California, 94080, USA
| | - Kalyani Penta
- Sutro Biopharma, Inc, 310 Utah Ave Ste 150, South San Francisco, California, 94080, USA
| | - Aaron K Sato
- Sutro Biopharma, Inc, 310 Utah Ave Ste 150, South San Francisco, California, 94080, USA
| | - Trevor J Hallam
- Sutro Biopharma, Inc, 310 Utah Ave Ste 150, South San Francisco, California, 94080, USA
| | - Gang Yin
- Sutro Biopharma, Inc, 310 Utah Ave Ste 150, South San Francisco, California, 94080, USA.
| |
Collapse
|
38
|
Bispecific antibody generated with sortase and click chemistry has broad antiinfluenza virus activity. Proc Natl Acad Sci U S A 2014; 111:16820-5. [PMID: 25385586 DOI: 10.1073/pnas.1408605111] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Bispecific antibodies have therapeutic potential by expanding the functions of conventional antibodies. Many different formats of bispecific antibodies have meanwhile been developed. Most are genetic modifications of the antibody backbone to facilitate incorporation of two different variable domains into a single molecule. Here, we present a bispecific format where we have fused two full-sized IgG antibodies via their C termini using sortase transpeptidation and click chemistry to create a covalently linked IgG antibody heterodimer. By linking two potent anti-influenza A antibodies together, we have generated a full antibody dimer with bispecific activity that retains the activity and stability of the two fusion partners.
Collapse
|
39
|
Schmohl L, Schwarzer D. Sortase-mediated ligations for the site-specific modification of proteins. Curr Opin Chem Biol 2014; 22:122-8. [PMID: 25299574 DOI: 10.1016/j.cbpa.2014.09.020] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 09/12/2014] [Accepted: 09/18/2014] [Indexed: 10/24/2022]
Abstract
Sortase-mediated ligation (SML) is one of the most commonly used techniques for the site-specific modification of proteins. Here, a brief overview on advantages and limitations of this technology in comparison with other chemoselective protein modification techniques is provided and successful approaches that extend the application range of SML are discussed.
Collapse
Affiliation(s)
- Lena Schmohl
- Interfaculty Institute of Biochemistry, University of Tuebingen, Hoppe-Seyler-Str. 4, D-72076 Tuebingen, Germany
| | - Dirk Schwarzer
- Interfaculty Institute of Biochemistry, University of Tuebingen, Hoppe-Seyler-Str. 4, D-72076 Tuebingen, Germany.
| |
Collapse
|
40
|
Heck T, Pham PH, Hammes F, Thöny-Meyer L, Richter M. Continuous Monitoring of Enzymatic Reactions on Surfaces by Real-Time Flow Cytometry: Sortase A Catalyzed Protein Immobilization as a Case Study. Bioconjug Chem 2014; 25:1492-500. [DOI: 10.1021/bc500230r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Tobias Heck
- Laboratory
for Bioactive Materials, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| | - Phu-Huy Pham
- Laboratory
for Bioactive Materials, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| | - Frederik Hammes
- Department
of Environmental Microbiology, Eawag, Swiss Federal Institute of Aquatic Science and Technology, Überlandstrasse 133, 8600 Dübendorf, Switzerland
| | - Linda Thöny-Meyer
- Laboratory
for Bioactive Materials, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| | - Michael Richter
- Laboratory
for Bioactive Materials, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| |
Collapse
|
41
|
Policarpo RL, Kang H, Liao X, Rabideau AE, Simon MD, Pentelute BL. Flow-based enzymatic ligation by sortase A. Angew Chem Int Ed Engl 2014; 53:9203-8. [PMID: 24989829 DOI: 10.1002/anie.201403582] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Indexed: 02/03/2023]
Abstract
Sortase-mediated ligation (sortagging) is a versatile, powerful strategy for protein modification. Because the sortase reaction reaches equilibrium, a large excess of polyglycine nucleophile is often employed to drive the reaction forward and suppress sortase-mediated side reactions. A flow-based sortagging platform employing immobilized sortase A within a microreactor was developed that permits efficient sortagging at low nucleophile concentrations. The platform was tested with several reaction partners and used to generate a protein bioconjugate inaccessible by solution-phase batch sortagging.
Collapse
Affiliation(s)
- Rocco L Policarpo
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 (USA)
| | | | | | | | | | | |
Collapse
|
42
|
Policarpo RL, Kang H, Liao X, Rabideau AE, Simon MD, Pentelute BL. Flow-Based Enzymatic Ligation by Sortase A. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201403582] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
43
|
Abstract
Bioorthogonal, chemoselective ligation methods are an essential part of the tools utilized to investigate biochemical pathways. Specifically enzymatic approaches are valuable methods in this context due to the inherent specificity of the deployed enzymes and the mild conditions of the modification reactions. One of the most common strategies is based on the transpeptidation catalyzed by sortase A derived from Staphylococcus aureus. The procedure is well established and a wide variety of applications have been published to date. Here, implementations of sortase A, which range from protein labeling using fluorescence dyes and the preparation of cyclic proteins to the modification of entire cells, are summarized. Furthermore, there is a focus on the optimization approaches established to solve the drawbacks of sortase-mediated transpeptidation.
Collapse
Affiliation(s)
- Markus Ritzefeld
- Bielefeld University, Department of Chemistry, Organic and Bioorganic Chemistry (OCIII), Universitätsstrasse 25, 33615 Bielefeld (Germany).
| |
Collapse
|
44
|
Liebscher S, Kornberger P, Fink G, Trost-Gross EM, Höss E, Skerra A, Bordusa F. Derivatization of antibody Fab fragments: a designer enzyme for native protein modification. Chembiochem 2014; 15:1096-100. [PMID: 24782039 DOI: 10.1002/cbic.201400059] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Indexed: 01/25/2023]
Abstract
Bioconjugates, such as antibody-drug conjugates, have gained recent attention because of their increasing use in therapeutic and diagnostic applications. Commonly used conjugation reactions based upon chemoselective reagents exhibit a number of drawbacks: most of these reactions lack regio- and stereospecificity, thus resulting in loss of protein functionality due to random modifications. Enzymes provide an obvious solution to this problem, but the intrinsic (natural) substrate specificities of existing enzymes pose severe limitations to the kind of modifications that can be introduced. Here we describe the application of the novel trypsin variant trypsiligase for site-specific modification of the C terminus of a Fab antibody fragment via a stable peptide bond. The suitability of this designed biocatalyst was demonstrated by coupling the Her2-specific Fab to artificial functionalities of either therapeutic (PEG) or diagnostic (fluorescein) relevance. In both cases we obtained homogeneously modified Fab products bearing the artificial functionality exclusively at the desired position.
Collapse
Affiliation(s)
- Sandra Liebscher
- Institute of Biochemistry/Biotechnology, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Strasse 3, 06120 Halle/Saale (Germany)
| | | | | | | | | | | | | |
Collapse
|
45
|
Li YM, Li YT, Pan M, Kong XQ, Huang YC, Hong ZY, Liu L. Irreversible site-specific hydrazinolysis of proteins by use of sortase. Angew Chem Int Ed Engl 2014; 53:2198-202. [PMID: 24470054 DOI: 10.1002/anie.201310010] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Indexed: 01/22/2023]
Abstract
Sortase-mediated hydrazinolysis of proteins with hydrazine or its derivatives was developed for the production of recombinant protein hydrazides. This process provides an alternative approach for protein semisynthesis through the use of recombinant protein hydrazides as thioester surrogates. It also provides an alternative method for C-terminal modification of proteins with functional units as well as for the preparation of C-to-C fusion proteins.
Collapse
Affiliation(s)
- Yi-Ming Li
- Tsinghua-Peking Center for Life Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing 100084 (China); School of Medical Engineering, Hefei University of Technology, Hefei, Anhui 230009 (China)
| | | | | | | | | | | | | |
Collapse
|
46
|
Li YM, Li YT, Pan M, Kong XQ, Huang YC, Hong ZY, Liu L. Irreversible Site-Specific Hydrazinolysis of Proteins by Use of Sortase. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201310010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
47
|
Heck T, Pham PH, Yerlikaya A, Thöny-Meyer L, Richter M. Sortase A catalyzed reaction pathways: a comparative study with six SrtA variants. Catal Sci Technol 2014. [DOI: 10.1039/c4cy00347k] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Different enzyme variants of sortase A fromStaphylococcus aureuswere found to have distinct catalytic properties with regard to site-directed protein fusion and competing intermolecular crosslinking reactions.
Collapse
Affiliation(s)
- Tobias Heck
- Empa
- Swiss Federal Laboratories for Materials Science and Technology
- Laboratory for Bioactive Materials
- 9014 St. Gallen, Switzerland
| | - Phu-Huy Pham
- Empa
- Swiss Federal Laboratories for Materials Science and Technology
- Laboratory for Bioactive Materials
- 9014 St. Gallen, Switzerland
| | - Alpaslan Yerlikaya
- Empa
- Swiss Federal Laboratories for Materials Science and Technology
- Laboratory for Bioactive Materials
- 9014 St. Gallen, Switzerland
| | - Linda Thöny-Meyer
- Empa
- Swiss Federal Laboratories for Materials Science and Technology
- Laboratory for Bioactive Materials
- 9014 St. Gallen, Switzerland
| | - Michael Richter
- Empa
- Swiss Federal Laboratories for Materials Science and Technology
- Laboratory for Bioactive Materials
- 9014 St. Gallen, Switzerland
| |
Collapse
|
48
|
Kornberger P, Skerra A. Sortase-catalyzed in vitro functionalization of a HER2-specific recombinant Fab for tumor targeting of the plant cytotoxin gelonin. MAbs 2013; 6:354-66. [PMID: 24492291 DOI: 10.4161/mabs.27444] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
We report on the preparation of a new type of immunotoxin via in vitro ligation of the αHer2 antigen binding fragment (Fab) of the clinically-validated antibody trastuzumab to the plant toxin gelonin, employing catalysis by the bacterial enzyme sortase A (SrtA). The αHer2 Fab was fused with the extended SrtA recognition motif LPET↓GLEH 6 at the C-terminus of its heavy chain, thereby preventing interference with antigen binding, while the toxin was equipped with a Gly 2 sequence at its N-terminus, distant to the catalytically active site in the C-terminal region. Site-specific in vitro transpeptidation led to a novel antibody-toxin conjugate wherein gelonin had effectively replaced the Fc region of a conventional (monomerized) immunoglobulin. After optimization of reaction conditions and incubation time, the resulting Fab-Gelonin ligation product was purified to homogeneity in a two-step procedure by means of Strep-Tactin affinity chromatography--utilizing the Strep-tag II appended to gelonin--and size exclusion chromatography. Binding activity of the immunotoxin for the Her2 ectodomain was indistinguishable from the unligated Fab as measured by real-time surface plasmon resonance spectroscopy. Specific cytotoxic potency of Fab-Gelonin was demonstrated against two Her2-positive cell lines, resulting in EC 50 values of ~1 nM or lower, indicating a 1000-fold enhanced cell-killing activity compared with gelonin itself. Thus, our strategy provides a convenient route to the modular construction of functional immunotoxins from Fabs of established tumor-specific antibodies with gelonin or related proteotoxins, also avoiding the elevated biosafety levels that would be mandatory for the direct biotechnological preparation of corresponding fusion proteins.
Collapse
Affiliation(s)
- Petra Kornberger
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie; Technische Universität München; Freising-Weihenstephan, Germany
| | - Arne Skerra
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie; Technische Universität München; Freising-Weihenstephan, Germany
| |
Collapse
|
49
|
Abstract
The process of protein crosslinking comprises the chemical, enzymatic, or chemoenzymatic formation of new covalent bonds between polypeptides. This allows (1) the site-directed coupling of proteins with distinct properties and (2) the de novo assembly of polymeric protein networks. Transferases, hydrolases, and oxidoreductases can be employed as catalysts for the synthesis of crosslinked proteins, thereby complementing chemical crosslinking strategies. Here, we review enzymatic approaches that are used for protein crosslinking at the industrial level or have shown promising potential in investigations on the lab-scale. We illustrate the underlying mechanisms of crosslink formation and point out the roles of the enzymes in their natural environments. Additionally, we discuss advantages and drawbacks of the enzyme-based crosslinking strategies and their potential for different applications.
Collapse
Affiliation(s)
- Tobias Heck
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Biomaterials, Lerchenfeldstrasse 5, CH-9014 St. Gallen, Switzerland
| | | | | | | |
Collapse
|
50
|
|