51
|
Steven J, Ubah OC, Buschhaus M, Kovaleva M, Ferguson L, Porter AJ, Barelle CJ. In Vitro Maturation of a Humanized Shark VNAR Domain to Improve Its Biophysical Properties. Methods Mol Biol 2020; 2070:115-142. [PMID: 31625093 DOI: 10.1007/978-1-4939-9853-1_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
VNAR domains are the binding regions of new antigen receptor proteins (IgNAR) which are unique to sharks, skates, and rays (Elasmobranchii). Individual VNAR domains can bind antigens independently and are the smallest reported adaptive immune recognition entities in the vertebrate kingdom. Sharing limited sequence homology with human immunoglobulin domains, their development and use as biotherapeutic agents require that they be humanized to minimize their potential immunogenicity. Efforts to humanize a human serum albumin (HSA)-specific VNAR, E06, resulted in protein molecules that initially had undesirable biophysical properties or reduced affinity for cognate antigen. Two lead humanized anti-HSA clones, v1.10 and v2.4, were subjected to a process of random mutagenesis using error-prone PCR. The mutated sequences for each humanized VNAR variant were screened for improvements in affinity for HSA and biophysical properties, achieved without a predicted increase in overall immunogenicity.
Collapse
Affiliation(s)
| | | | | | | | | | - Andrew J Porter
- Elasmogen Ltd., Aberdeen, UK
- Scottish Biologics Facility, School of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | | |
Collapse
|
52
|
Gebauer M, Skerra A. Engineering of binding functions into proteins. Curr Opin Biotechnol 2019; 60:230-241. [DOI: 10.1016/j.copbio.2019.05.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 05/07/2019] [Indexed: 12/13/2022]
|
53
|
Paloni JM, Dong XH, Olsen BD. Protein-Polymer Block Copolymer Thin Films for Highly Sensitive Detection of Small Proteins in Biological Fluids. ACS Sens 2019; 4:2869-2878. [PMID: 31702912 DOI: 10.1021/acssensors.9b01020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In nearly all biosensors, sensitivity is greatly reduced for measurements conducted in biological matrices due to nonspecific binding from off-target molecules. One method to overcome this issue is to design a sensor that enables selective size-based uptake of proteins. Herein, a protein-polymer conjugate thin-film biosensor is fabricated that self-assembles into lamellae containing alternating domains of protein and polymer. Analyte is captured in protein regions while polymer domains restrict diffusion of large molecules. Device sensitivity and size-based exclusion properties are probed using two analytes: streptavidin (SA, 52.8 kDa) and monomeric streptavidin (mSA2, 15.6 kDa). Tuning domain spacing by adjusting polymer molecular weight allows the design of films that relatively freely uptake mSA2 and largely restrict SA diffusion. Furthermore, when detecting the smaller mSA2, no reduction in the limit of detection (LOD) is observed when transitioning from detection in the buffer to detection in biological fluids. As a result, LOD measured in fluid samples is reduced by 2 orders of magnitude compared to a traditional surface-immobilized protein monolayer.
Collapse
Affiliation(s)
- Justin M. Paloni
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Xue-Hui Dong
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Bradley D. Olsen
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
54
|
Omersa N, Podobnik M, Anderluh G. Inhibition of Pore-Forming Proteins. Toxins (Basel) 2019; 11:E545. [PMID: 31546810 PMCID: PMC6784129 DOI: 10.3390/toxins11090545] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/27/2019] [Accepted: 09/10/2019] [Indexed: 12/16/2022] Open
Abstract
Perforation of cellular membranes by pore-forming proteins can affect cell physiology, tissue integrity, or immune response. Since many pore-forming proteins are toxins or highly potent virulence factors, they represent an attractive target for the development of molecules that neutralize their actions with high efficacy. There has been an assortment of inhibitors developed to specifically obstruct the activity of pore-forming proteins, in addition to vaccination and antibiotics that serve as a plausible treatment for the majority of diseases caused by bacterial infections. Here we review a wide range of potential inhibitors that can specifically and effectively block the activity of pore-forming proteins, from small molecules to more specific macromolecular systems, such as synthetic nanoparticles, antibodies, antibody mimetics, polyvalent inhibitors, and dominant negative mutants. We discuss their mechanism of inhibition, as well as advantages and disadvantages.
Collapse
Affiliation(s)
- Neža Omersa
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia.
| | - Marjetka Podobnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia.
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia.
| |
Collapse
|
55
|
Efficient evolved antibody mimetic designed ankyrin repeat proteins against programmed death-ligand 1 on E. coli surface display. Biochem Eng J 2019. [DOI: 10.1016/j.bej.2019.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
56
|
Fink M, Vazulka S, Egger E, Jarmer J, Grabherr R, Cserjan‐Puschmann M, Striedner G. Microbioreactor Cultivations of Fab‐ProducingEscherichia coliReveal Genome‐Integrated Systems as Suitable for Prospective Studies on Direct Fab Expression Effects. Biotechnol J 2019; 14:e1800637. [DOI: 10.1002/biot.201800637] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 06/11/2019] [Indexed: 11/07/2022]
Affiliation(s)
- Mathias Fink
- Christian Doppler Laboratory for Production of Next‐Level Biopharmaceuticals in E. coli, Department of BiotechnologyUniversity of Natural Resources and Life SciencesMuthgasse 18 A‐1190 Vienna Austria
| | - Sophie Vazulka
- Christian Doppler Laboratory for Production of Next‐Level Biopharmaceuticals in E. coli, Department of BiotechnologyUniversity of Natural Resources and Life SciencesMuthgasse 18 A‐1190 Vienna Austria
| | - Esther Egger
- Christian Doppler Laboratory for Production of Next‐Level Biopharmaceuticals in E. coli, Department of BiotechnologyUniversity of Natural Resources and Life SciencesMuthgasse 18 A‐1190 Vienna Austria
| | - Johanna Jarmer
- Boehringer Ingelheim RCV GmbH & Co KGDr. Boehringer‐Gasse 5‐11 A‐1120 Vienna Austria
| | - Reingard Grabherr
- Christian Doppler Laboratory for Production of Next‐Level Biopharmaceuticals in E. coli, Department of BiotechnologyUniversity of Natural Resources and Life SciencesMuthgasse 18 A‐1190 Vienna Austria
| | - Monika Cserjan‐Puschmann
- Christian Doppler Laboratory for Production of Next‐Level Biopharmaceuticals in E. coli, Department of BiotechnologyUniversity of Natural Resources and Life SciencesMuthgasse 18 A‐1190 Vienna Austria
| | - Gerald Striedner
- Christian Doppler Laboratory for Production of Next‐Level Biopharmaceuticals in E. coli, Department of BiotechnologyUniversity of Natural Resources and Life SciencesMuthgasse 18 A‐1190 Vienna Austria
| |
Collapse
|
57
|
Gao X, Conard A, Yang C, Zhan Y, Zeng F, Shi J, Li W, Dimitrov DS, Gong R. Optimization of the C-Terminus of an Autonomous Human IgG1 CH2 Domain for Stability and Aggregation Resistance. Mol Pharm 2019; 16:3647-3656. [DOI: 10.1021/acs.molpharmaceut.9b00544] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Xinyu Gao
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Alex Conard
- Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania 15261, United States
| | - Chunpeng Yang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yancheng Zhan
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fang Zeng
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jian Shi
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Wei Li
- Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania 15261, United States
| | - Dimiter S. Dimitrov
- Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania 15261, United States
| | - Rui Gong
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| |
Collapse
|
58
|
Pohlscheidt M, Kiss R, Gottschalk U. An Introduction to "Recent Trends in the Biotechnology Industry: Development and Manufacturing of Recombinant Antibodies and Proteins". ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2019; 165:1-8. [PMID: 29748871 DOI: 10.1007/10_2017_39] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The production of the first therapeutic proteins in the early 1980s heralded the launch of the biopharmaceuticals industry. The number of approved products has grown year on year over the past three decades to now represent a significant share of the entire pharmaceuticals market. More than 200 therapeutic proteins have been approved, approximately a quarter of which are represented by monoclonal antibodies and their derivatives. In 2016, the list of the top 15 best-selling drugs included more than eight biologics and in 2020 the trend will continue, with more than 50% of the top 20 best-selling drugs predicted to be biologics. From 1986 to 2014 several first-in-class, advance-in-class, and breakthrough designated therapeutic options were approved, with advanced therapies such as immuno-oncology and cell-based therapies being approved for several indications.
Collapse
Affiliation(s)
| | - Robert Kiss
- Biogen International GmbH, International Manufacturing, Zug, Switzerland
| | - Uwe Gottschalk
- Biogen International GmbH, International Manufacturing, Zug, Switzerland
| |
Collapse
|
59
|
Michel E, Plückthun A, Zerbe O. Peptide binding affinity redistributes preassembled repeat protein fragments. Biol Chem 2018; 400:395-404. [DOI: 10.1515/hsz-2018-0355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/21/2018] [Indexed: 01/21/2023]
Abstract
Abstract
Designed armadillo repeat proteins (dArmRPs) are modular peptide binders composed of N- and C-terminal capping repeats Y and A and a variable number of internal modules M that each specifically recognize two amino acids of the target peptide. Complementary fragments of dArmRPs obtained by splitting the protein between helices H1 and H2 of an internal module show conditional and specific assembly only in the presence of a target peptide (Michel, E., Plückthun, A., and Zerbe, O. (2018). Peptide-guided assembly of repeat protein fragments. Angew. Chem. Int. Ed. 57, 4576–4579). Here, we investigate dArmRP fragments that already spontaneously assemble with high affinity, e.g. those obtained from splits between entire modules or between helices H2 and H3. We find that the interaction of the peptide with the assembled fragments induces distal conformational rearrangements that suggest an induced fit on a global protein level. A population analysis of an equimolar mixture of an N-terminal and three C-terminal fragments with various affinities for the target peptide revealed predominant assembly of the weakest peptide binder. However, adding a target peptide to this mixture altered the population of the protein complexes such that the combination with the highest affinity for the peptide increased and becomes predominant when adding excess of peptide, highlighting the feasibility of peptide-induced enrichment of best binders from inter-modular fragment mixtures.
Collapse
Affiliation(s)
- Erich Michel
- Department of Chemistry , University of Zurich , Winterthurerstrasse 190 , CH-8057 Zürich , Switzerland
| | - Andreas Plückthun
- Department of Biochemistry , University of Zurich , Winterthurerstrasse 190 , CH-8057 Zürich , Switzerland
| | - Oliver Zerbe
- Department of Chemistry , University of Zurich , Winterthurerstrasse 190 , CH-8057 Zürich , Switzerland
| |
Collapse
|
60
|
Richards DA. Exploring alternative antibody scaffolds: Antibody fragments and antibody mimics for targeted drug delivery. DRUG DISCOVERY TODAY. TECHNOLOGIES 2018; 30:35-46. [PMID: 30553519 DOI: 10.1016/j.ddtec.2018.10.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/12/2018] [Accepted: 10/15/2018] [Indexed: 05/20/2023]
Abstract
The field of targeted therapeutics has benefitted immeasurably from the development of high-affinity antibodies. These important ligands have facilitated the development of effective therapies, particularly when conjugated to potent cytotoxic payloads i.e. in antibody-drug conjugates (ADCs). The success of ADCs is evidenced by rapid adoption within the pharmaceuticals community; many major companies have dedicated ADC research programmes. However, despite the advantages, the field of ADCs has failed to live up to its full potential. Studies have emerged suggesting that traditional IgG scaffolds may not be the optimal format for targeted payload delivery. In response, the protein engineering community has begun to explore alternative high-binding protein scaffolds as antibody mimics. In this short review I will summarise the generation, modification, and application of emerging antibody fragments and synthetic antibody mimics, with a focus on their use as drug carriers. The review aims to highlight the advantages of antibody mimics, and how they could be employed to overcome the issues and limitations of traditional ADCs.
Collapse
Affiliation(s)
- Daniel A Richards
- Department of Chemistry, University College London, 20 Gordon Street, London, WC1H 0AJ, UK.
| |
Collapse
|
61
|
Broad-Spectrum Antiviral Activity of an Ankyrin Repeat Protein on Viral Assembly against Chimeric NL4-3 Viruses Carrying Gag/PR Derived from Circulating Strains among Northern Thai Patients. Viruses 2018; 10:v10110625. [PMID: 30428529 PMCID: PMC6265948 DOI: 10.3390/v10110625] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 01/06/2023] Open
Abstract
Certain proteins have demonstrated proficient human immunodeficiency virus (HIV-1) life cycle disturbance. Recently, the ankyrin repeat protein targeting the HIV-1 capsid, AnkGAG1D4, showed a negative effect on the viral assembly of the HIV-1NL4-3 laboratory strain. To extend its potential for future clinical application, the activity of AnkGAG1D4 in the inhibition of other HIV-1 circulating strains was evaluated. Chimeric NL4-3 viruses carrying patient-derived Gag/PR-coding regions were generated from 131 antiretroviral drug-naïve HIV-1 infected individuals in northern Thailand during 2001–2012. SupT1, a stable T-cell line expressing AnkGAG1D4 and ankyrin non-binding control (AnkA32D3), were challenged with these chimeric viruses. The p24CA sequences were analysed and classified using the K-means clustering method. Among all the classes of virus classified using the p24CA sequences, SupT1/AnkGAG1D4 demonstrated significantly lower levels of p24CA than SupT1/AnkA32D3, which was found to correlate with the syncytia formation. This result suggests that AnkGAG1D4 can significantly interfere with the chimeric viruses derived from patients with different sequences of the p24CA domain. It supports the possibility of ankyrin-based therapy as a broad alternative therapeutic molecule for HIV-1 gene therapy in the future.
Collapse
|
62
|
Sokalingam S, Munussami G, Kim JR, Lee SG. Validation on the molecular docking efficiency of lipocalin family of proteins. J IND ENG CHEM 2018. [DOI: 10.1016/j.jiec.2018.06.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
63
|
Guo J, Luan X, Cong Z, Sun Y, Wang L, McKenna SL, Cahill MR, O'Driscoll CM. The potential for clinical translation of antibody-targeted nanoparticles in the treatment of acute myeloid leukaemia. J Control Release 2018; 286:154-166. [DOI: 10.1016/j.jconrel.2018.07.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 02/06/2023]
|
64
|
Awwad S, Angkawinitwong U. Overview of Antibody Drug Delivery. Pharmaceutics 2018; 10:E83. [PMID: 29973504 PMCID: PMC6161251 DOI: 10.3390/pharmaceutics10030083] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/29/2018] [Accepted: 06/29/2018] [Indexed: 12/11/2022] Open
Abstract
Monoclonal antibodies (mAbs) are one of the most important classes of therapeutic proteins, which are used to treat a wide number of diseases (e.g., oncology, inflammation and autoimmune diseases). Monoclonal antibody technologies are continuing to evolve to develop medicines with increasingly improved safety profiles, with the identification of new drug targets being one key barrier for new antibody development. There are many opportunities for developing antibody formulations for better patient compliance, cost savings and lifecycle management, e.g., subcutaneous formulations. However, mAb-based medicines also have limitations that impact their clinical use; the most prominent challenges are their short pharmacokinetic properties and stability issues during manufacturing, transport and storage that can lead to aggregation and protein denaturation. The development of long acting protein formulations must maintain protein stability and be able to deliver a large enough dose over a prolonged period. Many strategies are being pursued to improve the formulation and dosage forms of antibodies to improve efficacy and to increase the range of applications for the clinical use of mAbs.
Collapse
Affiliation(s)
- Sahar Awwad
- UCL School of Pharmacy, London WC1N 1AX, UK.
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1 V9EL, UK.
| | | |
Collapse
|
65
|
Winkler J. Extrahepatic Targeting of Oligonucleotides with Receptor-Binding Non-Immunoglobulin Scaffold Proteins. Nucleic Acid Ther 2018; 28:137-145. [PMID: 29733239 DOI: 10.1089/nat.2017.0713] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Although recent clinical successes of antisense, splice-switching, and siRNA oligonucleotides have established the therapeutic utility of this novel class of medicines, the efficient systemic application for non-liver targets remains elusive. Exploitation of active receptor-mediated targeting followed by efficient and productive cellular uptake is required for enabling the therapy of extrahepatic diseases on the expressional level. Evasion of liver accumulation and organ-specific targeting and also efficient cytosolic delivery after endosomal internalization are currently insufficiently solved issues. Lipid and polymer-based nanoparticles can be engineered for efficient cellular uptake and enhancement of endosomal escape, but are characterized by preferential liver accumulation based on biodistribution largely determined by particle size and biophysical properties. Oligonucleotide bioconjugates with receptor-binding ligands have been evolved for highly efficient targeting, but frequently result in a large extent of endosomal entrapment and consequently a lack of sufficient cytosolic concentrations. Non-immunoglobulin protein-based receptor recognition affords high cell-type selectivity and is promising for achieving nonhepatic oligonucleotide targeting. The use of such novel protein scaffolds, including designed ankyrin repeat proteins (DARPins), for oligonucleotide delivery is attractive for achieving effective tissue targeting. Issues for further development and optimization to advance approaches for extrahepatic oligonucleotide delivery by nanoparticles or bioconjugates are discussed.
Collapse
Affiliation(s)
- Johannes Winkler
- Department of Cardiology, Medical University of Vienna , Vienna, Austria
| |
Collapse
|
66
|
Setiawan D, Brender J, Zhang Y. Recent advances in automated protein design and its future challenges. Expert Opin Drug Discov 2018; 13:587-604. [PMID: 29695210 DOI: 10.1080/17460441.2018.1465922] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Protein function is determined by protein structure which is in turn determined by the corresponding protein sequence. If the rules that cause a protein to adopt a particular structure are understood, it should be possible to refine or even redefine the function of a protein by working backwards from the desired structure to the sequence. Automated protein design attempts to calculate the effects of mutations computationally with the goal of more radical or complex transformations than are accessible by experimental techniques. Areas covered: The authors give a brief overview of the recent methodological advances in computer-aided protein design, showing how methodological choices affect final design and how automated protein design can be used to address problems considered beyond traditional protein engineering, including the creation of novel protein scaffolds for drug development. Also, the authors address specifically the future challenges in the development of automated protein design. Expert opinion: Automated protein design holds potential as a protein engineering technique, particularly in cases where screening by combinatorial mutagenesis is problematic. Considering solubility and immunogenicity issues, automated protein design is initially more likely to make an impact as a research tool for exploring basic biology in drug discovery than in the design of protein biologics.
Collapse
Affiliation(s)
- Dani Setiawan
- a Department of Computational Medicine and Bioinformatics , University of Michigan , Ann Arbor , MI , USA
| | - Jeffrey Brender
- b Radiation Biology Branch , Center for Cancer Research, National Cancer Institute - NIH , Bethesda , MD , USA
| | - Yang Zhang
- a Department of Computational Medicine and Bioinformatics , University of Michigan , Ann Arbor , MI , USA.,c Department of Biological Chemistry , University of Michigan , Ann Arbor , MI , USA
| |
Collapse
|
67
|
Pranzatelli MR. Advances in Biomarker-Guided Therapy for Pediatric- and Adult-Onset Neuroinflammatory Disorders: Targeting Chemokines/Cytokines. Front Immunol 2018; 9:557. [PMID: 29670611 PMCID: PMC5893838 DOI: 10.3389/fimmu.2018.00557] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 03/05/2018] [Indexed: 12/26/2022] Open
Abstract
The concept and recognized components of “neuroinflammation” are expanding at the intersection of neurobiology and immunobiology. Chemokines (CKs), no longer merely necessary for immune cell trafficking and positioning, have multiple physiologic, developmental, and modulatory functionalities in the central nervous system (CNS) through neuron–glia interactions and other mechanisms affecting neurotransmission. They issue the “help me” cry of neurons and astrocytes in response to CNS injury, engaging invading lymphoid cells (T cells and B cells) and myeloid cells (dendritic cells, monocytes, and neutrophils) (adaptive immunity), as well as microglia and macrophages (innate immunity), in a cascade of events, some beneficial (reparative), others destructive (excitotoxic). Human cerebrospinal fluid (CSF) studies have been instrumental in revealing soluble immunobiomarkers involved in immune dysregulation, their dichotomous effects, and the cells—often subtype specific—that produce them. CKs/cytokines continue to be attractive targets for the pharmaceutical industry with varying therapeutic success. This review summarizes the developing armamentarium, complexities of not compromising surveillance/physiologic functions, and insights on applicable strategies for neuroinflammatory disorders. The main approach has been using a designer monoclonal antibody to bind directly to the chemo/cytokine. Another approach is soluble receptors to bind the chemo/cytokine molecule (receptor ligand). Recombinant fusion proteins combine a key component of the receptor with IgG1. An additional approach is small molecule antagonists (protein therapeutics, binding proteins, and protein antagonists). CK neutralizing molecules (“neutraligands”) that are not receptor antagonists, high-affinity neuroligands (“decoy molecules”), as well as neutralizing “nanobodies” (single-domain camelid antibody fragment) are being developed. Simultaneous, more precise targeting of more than one cytokine is possible using bispecific agents (fusion antibodies). It is also possible to inhibit part of a signaling cascade to spare protective cytokine effects. “Fusokines” (fusion of two cytokines or a cytokine and CK) allow greater synergistic bioactivity than individual cytokines. Another promising approach is experimental targeting of the NLRP3 inflammasome, amply expressed in the CNS and a key contributor to neuroinflammation. Serendipitous discovery is not to be discounted. Filling in knowledge gaps between pediatric- and adult-onset neuroinflammation by systematic collection of CSF data on CKs/cytokines in temporal and clinical contexts and incorporating immunobiomarkers in clinical trials is a challenge hereby set forth for clinicians and researchers.
Collapse
Affiliation(s)
- Michael R Pranzatelli
- National Pediatric Neuroinflammation Organization, Inc., Orlando, FL, United States.,College of Medicine, University of Central Florida, Orlando, FL, United States
| |
Collapse
|
68
|
Abou-Elkacem L, Wang H, Chowdhury SM, Kimura RH, Bachawal SV, Gambhir SS, Tian L, Willmann JK. Thy1-Targeted Microbubbles for Ultrasound Molecular Imaging of Pancreatic Ductal Adenocarcinoma. Clin Cancer Res 2018; 24:1574-1585. [PMID: 29301827 PMCID: PMC5884723 DOI: 10.1158/1078-0432.ccr-17-2057] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 11/09/2017] [Accepted: 12/18/2017] [Indexed: 12/17/2022]
Abstract
Purpose: To engineer a dual human and murine Thy1-binding single-chain-antibody ligand (Thy1-scFv) for contrast microbubble-enhanced ultrasound molecular imaging of pancreatic ductal adenocarcinoma (PDAC).Experimental Design: Thy1-scFv were engineered using yeast-surface-display techniques. Binding to soluble human and murine Thy1 and to Thy1-expressing cells was assessed by flow cytometry. Thy1-scFv was then attached to gas-filled microbubbles to create MBThy1-scFv Thy1 binding of MBThy1-scFv to Thy1-expressing cells was evaluated under flow shear stress conditions in flow-chamber experiments. MBscFv-scrambled and MBNon-targeted were used as negative controls. All microbubble types were tested in both orthotopic human PDAC xenografts and transgenic PDAC mice in vivoResults: Thy1-scFv had a KD of 3.4 ± 0.36 nmol/L for human and 9.2 ± 1.7 nmol/L for murine Thy1 and showed binding to both soluble and cellularly expressed Thy1. MBThy1-scFv was attached to Thy1 with high affinity compared with negative control microbubbles (P < 0.01) as assessed by flow cytometry. Similarly, flow-chamber studies showed significantly (P < 0.01) higher binding of MBThy1-scFv (3.0 ± 0.81 MB/cell) to Thy1-expressing cells than MBscFv-scrambled (0.57 ± 0.53) and MBNon-targeted (0.43 ± 0.53). In vivo ultrasound molecular imaging using MBThy1-scFv demonstrated significantly higher signal (P < 0.01) in both orthotopic (5.32 ± 1.59 a.u.) and transgenic PDAC (5.68 ± 2.5 a.u.) mice compared with chronic pancreatitis (0.84 ± 0.6 a.u.) and normal pancreas (0.67 ± 0.71 a.u.). Ex vivo immunofluorescence confirmed significantly (P < 0.01) increased Thy1 expression in PDAC compared with chronic pancreatitis and normal pancreas tissue.Conclusions: A dual human and murine Thy1-binding scFv was designed to generate contrast microbubbles to allow PDAC detection with ultrasound. Clin Cancer Res; 24(7); 1574-85. ©2018 AACR.
Collapse
Affiliation(s)
- Lotfi Abou-Elkacem
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford, California.
| | - Huaijun Wang
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford, California
| | - Sayan M Chowdhury
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford, California
| | - Richard H Kimura
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford, California
| | - Sunitha V Bachawal
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford, California
| | - Sanjiv S Gambhir
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford, California
| | - Lu Tian
- Department of Health, Research and Policy, Stanford University, Stanford, California
| | - Jürgen K Willmann
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford, California
| |
Collapse
|
69
|
Schiefner A, Gebauer M, Richter A, Skerra A. Anticalins Reveal High Plasticity in the Mode of Complex Formation with a Common Tumor Antigen. Structure 2018. [DOI: 10.1016/j.str.2018.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
70
|
Doytchinova IA, Flower DR. In silico prediction of cancer immunogens: current state of the art. BMC Immunol 2018; 19:11. [PMID: 29544447 PMCID: PMC5856276 DOI: 10.1186/s12865-018-0248-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 03/06/2018] [Indexed: 01/22/2023] Open
Abstract
Cancer kills 8 million annually worldwide. Although survival rates in prevalent cancers continue to increase, many cancers have no effective treatment, prompting the search for new and improved protocols. Immunotherapy is a new and exciting addition to the anti-cancer arsenal. The successful and accurate identification of aberrant host proteins acting as antigens for vaccination and immunotherapy is a key aspiration for both experimental and computational research. Here we describe key elements of in silico prediction, including databases of cancer antigens and bleeding-edge methodology for their prediction. We also highlight the role dendritic cell vaccines can play and how they can act as delivery mechanisms for epitope ensemble vaccines. Immunoinformatics can help streamline the discovery and utility of Cancer Immunogens.
Collapse
Affiliation(s)
- Irini A. Doytchinova
- Faculty of Pharmacy, Medical University of Sofia, 2 Dunav st, 1000 Sofia, Bulgaria
| | - Darren R. Flower
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, B4 7ET UK
| |
Collapse
|
71
|
Michel E, Plückthun A, Zerbe O. Peptide‐Guided Assembly of Repeat Protein Fragments. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201713377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Erich Michel
- Department of Chemistry University of Zurich Winterthurerstrasse 190 8057 Zurich Switzerland
| | - Andreas Plückthun
- Department of Biochemistry University of Zurich Winterthurerstrasse 190 8057 Zurich Switzerland
| | - Oliver Zerbe
- Department of Chemistry University of Zurich Winterthurerstrasse 190 8057 Zurich Switzerland
| |
Collapse
|
72
|
Michel E, Plückthun A, Zerbe O. Peptide-Guided Assembly of Repeat Protein Fragments. Angew Chem Int Ed Engl 2018; 57:4576-4579. [DOI: 10.1002/anie.201713377] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Erich Michel
- Department of Chemistry; University of Zurich; Winterthurerstrasse 190 8057 Zurich Switzerland
| | - Andreas Plückthun
- Department of Biochemistry; University of Zurich; Winterthurerstrasse 190 8057 Zurich Switzerland
| | - Oliver Zerbe
- Department of Chemistry; University of Zurich; Winterthurerstrasse 190 8057 Zurich Switzerland
| |
Collapse
|
73
|
Shirian J, Arkadash V, Cohen I, Sapir T, Radisky ES, Papo N, Shifman JM. Converting a broad matrix metalloproteinase family inhibitor into a specific inhibitor of MMP-9 and MMP-14. FEBS Lett 2018; 592:1122-1134. [PMID: 29473954 DOI: 10.1002/1873-3468.13016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 02/11/2018] [Accepted: 02/19/2018] [Indexed: 12/22/2022]
Abstract
MMP-14 and MMP-9 are two well-established cancer targets for which no specific clinically relevant inhibitor is available. Using a powerful combination of computational design and yeast surface display technology, we engineered such an inhibitor starting from a nonspecific MMP inhibitor, N-TIMP2. The engineered purified N-TIMP2 variants showed enhanced specificity toward MMP-14 and MMP-9 relative to a panel of off-target MMPs. MMP-specific N-TIMP2 sequence signatures were obtained that could be understood from the structural perspective of MMP/N-TIMP2 interactions. Our MMP-9 inhibitor exhibited 1000-fold preference for MMP-9 vs. MMP-14, which is likely to translate into significant differences under physiological conditions. Our results provide new insights regarding evolution of promiscuous proteins and optimization strategies for design of inhibitors with single-target specificities.
Collapse
Affiliation(s)
- Jason Shirian
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Israel
| | - Valeria Arkadash
- Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Itay Cohen
- Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Tamila Sapir
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Israel
| | - Evette S Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, USA
| | - Niv Papo
- Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Julia M Shifman
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Israel
| |
Collapse
|
74
|
Case BA, Kruziki MA, Stern LA, Hackel BJ. Evaluation of affibody charge modification identified by synthetic consensus design in molecular PET imaging of epidermal growth factor receptor. MOLECULAR SYSTEMS DESIGN & ENGINEERING 2018; 3:171-182. [PMID: 31467687 PMCID: PMC6715147 DOI: 10.1039/c7me00095b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Tumor overexpression of epidermal growth factor receptor (EGFR) correlates to therapeutic response in select patient populations. Thus, molecular positron emission tomography (PET) imaging of EGFR could stratify responders versus non-responders. We previously demonstrated effectiveness of a "synthetic consensus" design principle to identify six neutralizing mutations within a 58-amino acid EGFR-targeted affibody domain. Herein, we extend the approach to identify additional neutralized variants that vary net charge from -2 to either -4 or +4 while retaining high affinity (1.6 ± 1.2 nM and 2.5 ± 0.7 nM), specific binding to EGFR, secondary structure, and stability (Tm = 68 °C and 59 °C). We radiolabeled the resultant collection of five charge variants with 64Cu and evaluated PET imaging performance in murine models with subcutaneously xenografted EGFRhigh and EGFRlow tumors. All variants exhibited good EGFRhigh tumor imaging as early as 1 h, with EA35S (+3/-5) achieving 7.7 ± 1.4 %ID/g tumor at 4 h with 1.5 ± 0.3%ID/g EGFRlow tumor, 34 ± 5 tumor:muscle and 12 ± 3 tumor:blood ratios. The positively charged EA62S mutant (+6/-2) exhibited 2.2-3.3-fold higher liver signal than the other variants (p<0.01). The EA68 variant with higher charge density was more stable to human and mouse serum than neutralized variants. In a comparison of radiometal chelators, 1,4,7-triazacyclononane,1-glutaric acid-4,7-acetic acid (NODAGA) exhibited superior physiological specificity to 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA). In total, these studies comparatively evaluated a set of EGFR-targeted affibodies varying in net charge and charge density, which revealed functional variations that are useful in engineering an ideal probe for translational studies.
Collapse
Affiliation(s)
- Brett A Case
- Department of Chemical Engineering and Materials Science, University of Minnesota - Twin Cities, Minneapolis, MN 55455
| | - Max A Kruziki
- Department of Chemical Engineering and Materials Science, University of Minnesota - Twin Cities, Minneapolis, MN 55455
| | - Lawrence A Stern
- Department of Chemical Engineering and Materials Science, University of Minnesota - Twin Cities, Minneapolis, MN 55455
| | - Benjamin J Hackel
- Department of Chemical Engineering and Materials Science, University of Minnesota - Twin Cities, Minneapolis, MN 55455
| |
Collapse
|
75
|
Abstract
Therapeutic antibodies constitute one of the fastest areas of growth in the field of biologic drugs. A molecular understanding of how antibodies interact with their target antigens is known as epitope mapping. The data provided by epitope mapping is extremely valuable in the process of antibody humanization, as well as in vaccine design. In many cases the epitope recognized by the antibody is a complex, discontinuous 3D conformational epitope. Mapping the interactions of an antibody to a conformational epitope is difficult by many standard approaches. X-ray crystallography is considered to be the gold standard of epitope mapping as it can provide a near atomic resolution model of the antibody-antigen interaction. An X-ray structure allows for inspection of specific antibody-antigen interactions, even in the case of complex conformational epitopes. The method described here can be adapted for structure determination and epitope mapping of any antibody fragment to a simple or complex antigen.
Collapse
Affiliation(s)
- Moeko Toride King
- Department of Chemistry, California State University Fresno, 2555 E San Ramon Ave, Fresno, CA, 93740, USA
| | - Cory L Brooks
- Department of Chemistry, California State University Fresno, 2555 E San Ramon Ave, Fresno, CA, 93740, USA.
| |
Collapse
|
76
|
Friedrich L, Kornberger P, Mendler CT, Multhoff G, Schwaiger M, Skerra A. Selection of an Anticalin® against the membrane form of Hsp70 via bacterial surface display and its theranostic application in tumour models. Biol Chem 2017; 399:235-252. [DOI: 10.1515/hsz-2017-0207] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/23/2017] [Indexed: 01/08/2023]
Abstract
Abstract
We describe the selection of Anticalins against a common tumour surface antigen, human Hsp70, using functional display on live Escherichia coli cells as fusion with a truncated EspP autotransporter. While found intracellularly in normal cells, Hsp70 is frequently exposed in a membrane-bound state on the surface of tumour cells and, even more pronounced, in metastases or after radiochemotherapy. Employing a recombinant Hsp70 fragment comprising residues 383-548 as the target, Anticalins were selected from a naïve bacterial library. The Anticalin with the highest affinity (K
D=13 nm), as determined towards recombinant full-length Hsp70 by real-time surface plasmon resonance analysis, was improved to K
D=510 pm by doped random mutagenesis and another cycle of E. coli surface display, followed by rational combination of mutations. This Anticalin, which recognises a linear peptide epitope located in the interdomain linker of Hsp70, was demonstrated to specifically bind Hsp70 in its membrane-associated form in immunofluorescence microscopy and via flow cytometry using the FaDu cell line, which is positive for surface Hsp70. The radiolabelled and PASylated Anticalin revealed specific tumour accumulation in xenograft mice using positron emission tomography (PET) imaging. Furthermore, after enzymatic coupling to the protein toxin gelonin, the Anticalin showed potent cytotoxicity on FaDu cells in vitro.
Collapse
Affiliation(s)
- Lars Friedrich
- Munich Center for Integrated Protein Science, CIPS-M, and Lehrstuhl für Biologische Chemie , Technische Universität München , D-85354 Freising (Weihenstephan) , Germany
| | - Petra Kornberger
- Munich Center for Integrated Protein Science, CIPS-M, and Lehrstuhl für Biologische Chemie , Technische Universität München , D-85354 Freising (Weihenstephan) , Germany
| | - Claudia T. Mendler
- Munich Center for Integrated Protein Science, CIPS-M, and Lehrstuhl für Biologische Chemie , Technische Universität München , D-85354 Freising (Weihenstephan) , Germany
| | - Gabriele Multhoff
- Department of Radiation Oncology , Klinikum rechts der Isar, Technische Universität München , D-81675 München , Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, Klinikum rechts der Isar , Technische Universität München , D-81675 München , Germany
| | - Arne Skerra
- Munich Center for Integrated Protein Science, CIPS-M, and Lehrstuhl für Biologische Chemie , Technische Universität München , D-85354 Freising (Weihenstephan) , Germany
| |
Collapse
|
77
|
Kalichuk V, Renodon-Cornière A, Béhar G, Carrión F, Obal G, Maillasson M, Mouratou B, Préat V, Pecorari F. A novel, smaller scaffold for Affitins: Showcase with binders specific for EpCAM. Biotechnol Bioeng 2017; 115:290-299. [DOI: 10.1002/bit.26463] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/17/2017] [Accepted: 09/25/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Valentina Kalichuk
- CRCINA, Inserm, CNRS, Université d'Angers; Université de Nantes; Nantes France
- Université Catholique de Louvain; Louvain Drug Research Institute; Advanced Drug Delivery and Biomaterials; Brussels Belgium
| | | | - Ghislaine Béhar
- CRCINA, Inserm, CNRS, Université d'Angers; Université de Nantes; Nantes France
| | - Federico Carrión
- Institut Pasteur de Montevideo; Protein Biophysics Unit; Montevideo Uruguay
| | - Gonzalo Obal
- Institut Pasteur de Montevideo; Protein Biophysics Unit; Montevideo Uruguay
| | - Mike Maillasson
- CRCINA, Inserm, CNRS, Université d'Angers; Université de Nantes; Nantes France
- Impact, CRCINA, Inserm, CNRS; Université d'Angers; Université de Nantes; Nantes France
| | - Barbara Mouratou
- CRCINA, Inserm, CNRS, Université d'Angers; Université de Nantes; Nantes France
| | - Véronique Préat
- Université Catholique de Louvain; Louvain Drug Research Institute; Advanced Drug Delivery and Biomaterials; Brussels Belgium
| | - Frédéric Pecorari
- CRCINA, Inserm, CNRS, Université d'Angers; Université de Nantes; Nantes France
| |
Collapse
|
78
|
Steven J, Müller MR, Carvalho MF, Ubah OC, Kovaleva M, Donohoe G, Baddeley T, Cornock D, Saunders K, Porter AJ, Barelle CJ. In Vitro Maturation of a Humanized Shark VNAR Domain to Improve Its Biophysical Properties to Facilitate Clinical Development. Front Immunol 2017; 8:1361. [PMID: 29109729 PMCID: PMC5660122 DOI: 10.3389/fimmu.2017.01361] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/04/2017] [Indexed: 11/13/2022] Open
Abstract
Molecular engineering to increase the percentage identity to common human immunoglobulin sequences of non-human therapeutic antibodies and scaffolds has become standard practice. This strategy is often used to reduce undesirable immunogenic responses, accelerating the clinical development of candidate domains. The first humanized shark variable domain (VNAR) was reported by Kovalenko and colleagues and used the anti-human serum albumin (HSA) domain, clone E06, as a model to construct a number of humanized versions including huE06v1.10. This study extends this work by using huE06v1.10 as a template to isolate domains with improved biophysical properties and reduced antigenicity. Random mutagenesis was conducted on huE06v1.10 followed by refinement of clones through an off-rate ranking-based selection on target antigen. Many of these next-generation binders retained high affinity for target, together with good species cross-reactivity. Lead domains were assessed for any tendency to dimerize, tolerance to N- and C-terminal fusions, affinity, stability, and relative antigenicity in human dendritic cell assays. Functionality of candidate clones was verified in vivo through the extension of serum half-life in a typical drug format. From these analyses the domain, BA11, exhibited negligible antigenicity, high stability and high affinity for mouse, rat, and HSA. When these attributes were combined with demonstrable functionality in a rat model of PK, the BA11 clone was established as our clinical candidate.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Thomas Baddeley
- Department of Chemistry, University of Aberdeen, Aberdeen, United Kingdom
| | - Dawn Cornock
- Department of Chemistry, University of Aberdeen, Aberdeen, United Kingdom
| | | | - Andrew J Porter
- Elasmogen Ltd., Aberdeen, United Kingdom.,Department of Molecular and Cell Biology, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | | |
Collapse
|
79
|
Chevalier A, Silva DA, Rocklin GJ, Hicks DR, Vergara R, Murapa P, Bernard SM, Zhang L, Lam KH, Yao G, Bahl CD, Miyashita SI, Goreshnik I, Fuller JT, Koday MT, Jenkins CM, Colvin T, Carter L, Bohn A, Bryan CM, Fernández-Velasco DA, Stewart L, Dong M, Huang X, Jin R, Wilson IA, Fuller DH, Baker D. Massively parallel de novo protein design for targeted therapeutics. Nature 2017; 550:74-79. [PMID: 28953867 PMCID: PMC5802399 DOI: 10.1038/nature23912] [Citation(s) in RCA: 278] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 08/17/2017] [Indexed: 12/24/2022]
Abstract
De novo protein design holds promise for creating small stable proteins with shapes customized to bind therapeutic targets. We describe a massively parallel approach for designing, manufacturing and screening mini-protein binders, integrating large-scale computational design, oligonucleotide synthesis, yeast display screening and next-generation sequencing. We designed and tested 22,660 mini-proteins of 37-43 residues that target influenza haemagglutinin and botulinum neurotoxin B, along with 6,286 control sequences to probe contributions to folding and binding, and identified 2,618 high-affinity binders. Comparison of the binding and non-binding design sets, which are two orders of magnitude larger than any previously investigated, enabled the evaluation and improvement of the computational model. Biophysical characterization of a subset of the binder designs showed that they are extremely stable and, unlike antibodies, do not lose activity after exposure to high temperatures. The designs elicit little or no immune response and provide potent prophylactic and therapeutic protection against influenza, even after extensive repeated dosing.
Collapse
Affiliation(s)
- Aaron Chevalier
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, USA
- Institute for Protein Design, University of Washington, Seattle, Washington 98195, USA
| | - Daniel-Adriano Silva
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, USA
- Institute for Protein Design, University of Washington, Seattle, Washington 98195, USA
| | - Gabriel J Rocklin
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, USA
- Institute for Protein Design, University of Washington, Seattle, Washington 98195, USA
| | - Derrick R Hicks
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, USA
- Institute for Protein Design, University of Washington, Seattle, Washington 98195, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, Washington 98195, USA
| | - Renan Vergara
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, USA
- Institute for Protein Design, University of Washington, Seattle, Washington 98195, USA
- Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Ciudad Universitaria, México City 04510, Mexico
| | - Patience Murapa
- Department of Microbiology, University of Washington, Seattle, Washington 98109, USA
| | - Steffen M Bernard
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Lu Zhang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian 350002, China
- Department of Chemistry and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Kwok-Ho Lam
- Department of Physiology and Biophysics, University of California, Irvine, California 92697, USA
| | - Guorui Yao
- Department of Physiology and Biophysics, University of California, Irvine, California 92697, USA
| | - Christopher D Bahl
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, USA
- Institute for Protein Design, University of Washington, Seattle, Washington 98195, USA
| | - Shin-Ichiro Miyashita
- Department of Urology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Microbiology and Immunobiology and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Inna Goreshnik
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, USA
| | - James T Fuller
- Department of Microbiology, University of Washington, Seattle, Washington 98109, USA
| | - Merika T Koday
- Department of Microbiology, University of Washington, Seattle, Washington 98109, USA
- Virvio Inc., Seattle, Washington 98195, USA
| | - Cody M Jenkins
- Department of Microbiology, University of Washington, Seattle, Washington 98109, USA
| | - Tom Colvin
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, USA
| | - Lauren Carter
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, USA
- Institute for Protein Design, University of Washington, Seattle, Washington 98195, USA
| | - Alan Bohn
- Department of Microbiology, University of Washington, Seattle, Washington 98109, USA
| | - Cassie M Bryan
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, USA
- Institute for Protein Design, University of Washington, Seattle, Washington 98195, USA
| | - D Alejandro Fernández-Velasco
- Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Ciudad Universitaria, México City 04510, Mexico
| | - Lance Stewart
- Institute for Protein Design, University of Washington, Seattle, Washington 98195, USA
| | - Min Dong
- Department of Urology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Microbiology and Immunobiology and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Xuhui Huang
- Department of Chemistry and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Rongsheng Jin
- Department of Physiology and Biophysics, University of California, Irvine, California 92697, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Deborah H Fuller
- Department of Microbiology, University of Washington, Seattle, Washington 98109, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, USA
- Institute for Protein Design, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
80
|
Zhang S, Geryak R, Geldmeier J, Kim S, Tsukruk VV. Synthesis, Assembly, and Applications of Hybrid Nanostructures for Biosensing. Chem Rev 2017; 117:12942-13038. [DOI: 10.1021/acs.chemrev.7b00088] [Citation(s) in RCA: 206] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Shuaidi Zhang
- School of Materials Science
and Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332-0245, United States
| | - Ren Geryak
- School of Materials Science
and Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332-0245, United States
| | - Jeffrey Geldmeier
- School of Materials Science
and Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332-0245, United States
| | - Sunghan Kim
- School of Materials Science
and Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332-0245, United States
| | - Vladimir V. Tsukruk
- School of Materials Science
and Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332-0245, United States
| |
Collapse
|
81
|
Binder U, Skerra A. PASylation®: A versatile technology to extend drug delivery. Curr Opin Colloid Interface Sci 2017. [DOI: 10.1016/j.cocis.2017.06.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
82
|
Zhu L, Zhao Z, Cheng P, He Z, Cheng Z, Peng J, Wang H, Wang C, Yang Y, Hu Z. Antibody-Mimetic Peptoid Nanosheet for Label-Free Serum-Based Diagnosis of Alzheimer's Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1700057. [PMID: 28605073 DOI: 10.1002/adma.201700057] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/28/2017] [Indexed: 06/07/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia characterized by progressive cognitive decline. Current diagnosis of AD is based on symptoms, neuropsychological tests, and neuroimaging, and is usually evident years after the pathological process. Early assessment at the preclinical or prodromal stage is in a great demand since treatment after the onset can hardly stop or reverse the disease progress. However, early diagnosis of AD is challenging due to the lack of reliable noninvasive approaches. Here, an antibody-mimetic self-assembling peptoid nanosheet containing surface-exposed Aβ42-recognizing loops is constructed, and a label-free sensor for the detection of AD serum is developed. The loop-displaying peptoid nanosheet is demonstrated to have high affinity to serum Aβ42, and to be able to identify AD sera with high sensitivity. The dense distribution of molecular recognition loops on the robust peptoid nanosheet scaffold not only mimics the architecture of antibodies, but also reduces the nonspecific binding in detecting multicomponent samples. This antibody-mimetic 2D material holds great potential toward the blood-based diagnosis of AD, and meanwhile provides novel insights into the antibody alternative engineering and the universal application in biological and chemical sensors.
Collapse
Affiliation(s)
- Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Zijian Zhao
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Peng Cheng
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Zhaohui He
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhiqiang Cheng
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jiaxi Peng
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Huayi Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Zhiyuan Hu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Sino-Danish Colloge, University of Chinese Academy of Sciences, Beijing, 100049, China
- Yangtze River Delta Academy of Nanotechnology and Industry Development Research, Zhejiang Province, Jiaxing, 314000, China
| |
Collapse
|
83
|
Stein V, Nabi M, Alexandrov K. Ultrasensitive Scaffold-Dependent Protease Sensors with Large Dynamic Range. ACS Synth Biol 2017; 6:1337-1342. [PMID: 28291337 DOI: 10.1021/acssynbio.6b00370] [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] [Indexed: 11/29/2022]
Abstract
The rational construction of synthetic protein switches with predefined input-output parameters constitutes a key goal of synthetic biology with many potential applications ranging from metabolic engineering to diagnostics. Yet, generally applicable strategies to construct tailor-engineered protein switches have so far remained elusive. Here, we use SpyTag/SpyCatcher-mediated protein ligation to engineer modularly organized, scaffold-dependent protease sensors that exploit a combination of affinity targeting and protease-inducible protein-protein interactions. We use this architecture to create a suite of integrated signal sensing and amplification circuits that can detect the activity of α-thrombin and prostate specific antigen with a dynamic range covering 5 orders of magnitude. We determine the key design features critical for signal transmission between protease-based sensors, transducers, and actuators.
Collapse
Affiliation(s)
- Viktor Stein
- Institute
for Molecular Biosciences, The University of Queensland, QBP Building 80, St Lucia, Queensland 4072, Australia
| | - Masuda Nabi
- Institute
for Molecular Biosciences, The University of Queensland, QBP Building 80, St Lucia, Queensland 4072, Australia
| | - Kirill Alexandrov
- Institute
for Molecular Biosciences, The University of Queensland, QBP Building 80, St Lucia, Queensland 4072, Australia
| |
Collapse
|
84
|
Kim MA, Yoon HS, Park SH, Kim DY, Pyo A, Kim HS, Min JJ, Hong Y. Engineering of monobody conjugates for human EphA2-specific optical imaging. PLoS One 2017; 12:e0180786. [PMID: 28686661 PMCID: PMC5501600 DOI: 10.1371/journal.pone.0180786] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 06/21/2017] [Indexed: 02/06/2023] Open
Abstract
In a previous study, we developed an E1 monobody specific for the tumor biomarker hEphA2 [PLoS ONE (2015) 10(7): e0132976]. E1 showed potential as a molecular probe for in vitro and in vivo targeting of cancers overexpressing hEphA2. In the present study, we constructed expression vectors for E1 conjugated to optical reporters such as Renilla luciferase variant 8 (Rluc8) or enhanced green fluorescent protein (EGFP) and purified such recombinant proteins by affinity chromatography in E. coli. E1-Rluc8 and E1-EGFP specifically bound to hEphA2 in human prostate cancer PC3 cells but not in human cervical cancer HeLa cells, which express hEphA2 at high and low levels, respectively. These recombinant proteins maintained >40% activity in mouse serum at 24 h. In vivo optical imaging for 24 h did not detect E1-EGFP signals, whereas E1-Rluc8 showed tumor-specific luminescence signals in PC3 but not in HeLa xenograft mice. E1-Rluc8 signals were detected at 4 h, peaked at 12 h, and were undetectable at 24 h. These results suggest the potential of E1-Rluc8 as an EphA2-specific optical imaging agent.
Collapse
MESH Headings
- Animals
- Antibodies, Neoplasm/biosynthesis
- Antibodies, Neoplasm/chemistry
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Line, Tumor
- Escherichia coli/genetics
- Escherichia coli/metabolism
- Female
- Genes, Reporter
- Genetic Vectors/chemistry
- Genetic Vectors/metabolism
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- HeLa Cells
- Heterografts
- Humans
- Immunoconjugates/chemistry
- Immunoconjugates/metabolism
- Luciferases/genetics
- Luciferases/metabolism
- Male
- Mice, Inbred BALB C
- Mice, Nude
- Optical Imaging
- Organ Specificity
- Protein Engineering
- Receptor, EphA2/analysis
- Receptor, EphA2/genetics
- Receptor, EphA2/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
Collapse
Affiliation(s)
- Min-A Kim
- Department of Microbiology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Department of Molecular Medicine (BK21Plus), Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Hee Seung Yoon
- Department of Molecular Medicine (BK21Plus), Chonnam National University Medical School, Gwangju, Republic of Korea
- Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Seung-Hwan Park
- Biological Resource Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Jeongeup, Republic of Korea
| | - Dong-Yeon Kim
- Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Ayoung Pyo
- Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Hyeon Sik Kim
- Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Jung-Joon Min
- Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Yeongjin Hong
- Department of Microbiology, Chonnam National University Medical School, Gwangju, Republic of Korea
- * E-mail:
| |
Collapse
|
85
|
Tools and limitations to study the molecular composition of synapses by fluorescence microscopy. Biochem J 2017; 473:3385-3399. [PMID: 27729584 DOI: 10.1042/bcj20160366] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 06/23/2016] [Indexed: 01/21/2023]
Abstract
The synapse is densely packed with proteins involved in various highly regulated processes. Synaptic protein copy numbers and their stoichiometric distribution have a drastic influence on neuronal integrity and function. Therefore, the molecular analysis of synapses is a key element to understand their architecture and function. The overall structure of the synapse has been revealed with an exquisite amount of details by electron microscopy. However, the molecular composition and the localization of proteins are more easily addressed with fluorescence imaging, especially with the improved resolution achieved by super-resolution microscopy techniques. Notably, the fast improvement of imaging instruments has not been reflected in the optimization of biological sample preparation. During recent years, large efforts have been made to generate affinity probes smaller than conventional antibodies adapted for fluorescent super-resolution imaging. In this review, we briefly discuss the current views on synaptic organization and necessary key technologies to progress in the understanding of synaptic physiology. We also highlight the challenges faced by current fluorescent super-resolution methods, and we describe the prerequisites for an ideal study of synaptic organization.
Collapse
|
86
|
Yu X, Yang YP, Dikici E, Deo SK, Daunert S. Beyond Antibodies as Binding Partners: The Role of Antibody Mimetics in Bioanalysis. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2017; 10:293-320. [PMID: 28375702 PMCID: PMC5895458 DOI: 10.1146/annurev-anchem-061516-045205] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The emergence of novel binding proteins or antibody mimetics capable of binding to ligand analytes in a manner analogous to that of the antigen-antibody interaction has spurred increased interest in the biotechnology and bioanalytical communities. The goal is to produce antibody mimetics designed to outperform antibodies with regard to binding affinities, cellular and tumor penetration, large-scale production, and temperature and pH stability. The generation of antibody mimetics with tailored characteristics involves the identification of a naturally occurring protein scaffold as a template that binds to a desired ligand. This scaffold is then engineered to create a superior binder by first creating a library that is then subjected to a series of selection steps. Antibody mimetics have been successfully used in the development of binding assays for the detection of analytes in biological samples, as well as in separation methods, cancer therapy, targeted drug delivery, and in vivo imaging. This review describes recent advances in the field of antibody mimetics and their applications in bioanalytical chemistry, specifically in diagnostics and other analytical methods.
Collapse
Affiliation(s)
- Xiaowen Yu
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida 33136;
| | - Yu-Ping Yang
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida 33136;
| | - Emre Dikici
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida 33136;
| | - Sapna K Deo
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida 33136;
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida 33136;
| |
Collapse
|
87
|
Ståhl S, Gräslund T, Eriksson Karlström A, Frejd FY, Nygren PÅ, Löfblom J. Affibody Molecules in Biotechnological and Medical Applications. Trends Biotechnol 2017; 35:691-712. [PMID: 28514998 DOI: 10.1016/j.tibtech.2017.04.007] [Citation(s) in RCA: 243] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/18/2017] [Accepted: 04/20/2017] [Indexed: 01/08/2023]
Abstract
Affibody molecules are small (6.5-kDa) affinity proteins based on a three-helix bundle domain framework. Since their introduction 20 years ago as an alternative to antibodies for biotechnological applications, the first therapeutic affibody molecules have now entered clinical development and more than 400 studies have been published in which affibody molecules have been developed and used in a variety of contexts. In this review, we focus primarily on efforts over the past 5 years to explore the potential of affibody molecules for medical applications in oncology, neurodegenerative, and inflammation disorders, including molecular imaging, receptor signal blocking, and delivery of toxic payloads. In addition, we describe recent examples of biotechnological applications, in which affibody molecules have been exploited as modular affinity fusion partners.
Collapse
Affiliation(s)
- Stefan Ståhl
- Division of Protein Technology, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden.
| | - Torbjörn Gräslund
- Division of Protein Technology, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| | | | - Fredrik Y Frejd
- Unit of Biomedical Radiation Sciences, Uppsala University, SE-751 85 Uppsala, Sweden; Affibody AB, Gunnar Asplunds Allé 24, SE-171 69 Solna, Sweden
| | - Per-Åke Nygren
- Division of Protein Technology, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| | - John Löfblom
- Division of Protein Technology, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| |
Collapse
|
88
|
Pitalúa-Cortés QG, García-Pérez FO, Villaseñor-Navarro Y, Lara-Medina FU, Matus-Santos JA, Soldevilla-Gallardo I, Porras-Reyes FI, Pérez-Sánchez VM, Maldonado-Martínez HA, Pérez-Báez W, Sollozo-Dupont I. 68Ga-DTPA Anti-HER2 positron emission tomography/CT successfully predicts the overexpression of human epidermal growth factor receptor in lung metastases from breast cancer. BJR Case Rep 2017; 3:20160136. [PMID: 30363270 PMCID: PMC6159203 DOI: 10.1259/bjrcr.20160136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 03/10/2017] [Accepted: 03/22/2017] [Indexed: 12/19/2022] Open
Abstract
Molecular identification of a metastatic tumour without the inconvenience of a biopsy and the time required for pathological characterization is possible using molecular imaging. Here, we present the case of a patient with breast cancer in whom 68Ga-diethylenetriamine pentaacetic acid anti-human epidermal growth factor receptor 2 positron emission tomography-CT was successfully employed to characterize the expression of human epidermal growth factor receptor 2 in metastatic sites.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Wendy Pérez-Báez
- Deparment of Surgical Pathology, Instituto Nacional de Cancerología, INCan, México
| | | |
Collapse
|
89
|
Haußner C, Damm D, Nirschl S, Rohrhofer A, Schmidt B, Eichler J. Peptide Paratope Mimics of the Broadly Neutralizing HIV-1 Antibody b12. Chembiochem 2017; 18:647-653. [DOI: 10.1002/cbic.201600621] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Christina Haußner
- Department of Chemistry and Pharmacy; University of Erlangen-Nürnberg; Schuhstrasse 19 91052 Erlangen Germany
| | - Dominik Damm
- Institute of Medical Microbiology and Hygiene; University of Regensburg; Franz-Josef-Strauss-Allee 11 93053 Regensburg Germany
| | - Sandra Nirschl
- Institute of Medical Microbiology and Hygiene; University of Regensburg; Franz-Josef-Strauss-Allee 11 93053 Regensburg Germany
| | - Anette Rohrhofer
- Institute of Clinical Microbiology and Hygiene; University of Regensburg; Franz-Josef-Strauss-Allee 11 93053 Regensburg Germany
| | - Barbara Schmidt
- Institute of Medical Microbiology and Hygiene; University of Regensburg; Franz-Josef-Strauss-Allee 11 93053 Regensburg Germany
- Institute of Clinical Microbiology and Hygiene; University of Regensburg; Franz-Josef-Strauss-Allee 11 93053 Regensburg Germany
| | - Jutta Eichler
- Department of Chemistry and Pharmacy; University of Erlangen-Nürnberg; Schuhstrasse 19 91052 Erlangen Germany
| |
Collapse
|
90
|
Li YF, Sun YM, Beier RC, Lei HT, Gee S, Hammock BD, Wang H, Wang Z, Sun X, Shen YD, Yang JY, Xu ZL. Immunochemical techniques for multianalyte analysis of chemical residues in food and the environment: A review. Trends Analyt Chem 2017. [DOI: 10.1016/j.trac.2016.12.010] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
91
|
Coppock MB, Warner CR, Dorsey B, Orlicki JA, Sarkes DA, Lai BT, Pitram SM, Rohde RD, Malette J, Wilson JA, Kearney P, Fang KC, Law SM, Candelario SL, Farrow B, Finch AS, Agnew HD, Heath JR, Stratis‐Cullum DN. Protein catalyzed capture agents with tailored performance for in vitro and in vivo applications. Biopolymers 2017; 108:e22934. [PMID: 27539157 PMCID: PMC6585716 DOI: 10.1002/bip.22934] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/25/2016] [Accepted: 08/17/2016] [Indexed: 12/12/2022]
Abstract
We report on peptide-based ligands matured through the protein catalyzed capture (PCC) agent method to tailor molecular binders for in vitro sensing/diagnostics and in vivo pharmacokinetics parameters. A vascular endothelial growth factor (VEGF) binding peptide and a peptide against the protective antigen (PA) protein of Bacillus anthracis discovered through phage and bacterial display panning technologies, respectively, were modified with click handles and subjected to iterative in situ click chemistry screens using synthetic peptide libraries. Each azide-alkyne cycloaddition iteration, promoted by the respective target proteins, yielded improvements in metrics for the application of interest. The anti-VEGF PCC was explored as a stable in vivo imaging probe. It exhibited excellent stability against proteases and a mean elimination in vivo half-life (T1/2 ) of 36 min. Intraperitoneal injection of the reagent results in slow clearance from the peritoneal cavity and kidney retention at extended times, while intravenous injection translates to rapid renal clearance. The ligand competed with the commercial antibody for binding to VEGF in vivo. The anti-PA ligand was developed for detection assays that perform in demanding physical environments. The matured anti-PA PCC exhibited no solution aggregation, no fragmentation when heated to 100°C, and > 81% binding activity for PA after heating at 90°C for 1 h. We discuss the potential of the PCC agent screening process for the discovery and enrichment of next generation antibody alternatives.
Collapse
Affiliation(s)
- Matthew B. Coppock
- Sensors and Electron Devices DirectorateU.S. Army Research LaboratoryAdelphiMD20783
| | - Candice R. Warner
- Excet, SpringfieldVA 22151 supporting USA Edgewood Chemical Biological CenterAberdeen Proving GroundMD21010
| | - Brandi Dorsey
- Federal Staffing Resources, Annapolis, MD supporting U.S. Army Research LaboratoryAdelphiMD20783
| | - Joshua A. Orlicki
- Weapons and Materials Research DirectorateU.S. Army Research LaboratoryAberdeen Proving GroundMD21005
| | - Deborah A. Sarkes
- Sensors and Electron Devices DirectorateU.S. Army Research LaboratoryAdelphiMD20783
| | - Bert T. Lai
- Indi Molecular6162 Bristol ParkwayCulver CityCA90230
| | | | | | | | | | | | | | | | | | - Blake Farrow
- Division of Chemistry and Chemical EngineeringCalifornia Institute of Technology1200 East California BoulevardPasadenaCA91125
| | - Amethist S. Finch
- Sensors and Electron Devices DirectorateU.S. Army Research LaboratoryAdelphiMD20783
| | | | - James R. Heath
- Division of Chemistry and Chemical EngineeringCalifornia Institute of Technology1200 East California BoulevardPasadenaCA91125
| | | |
Collapse
|
92
|
In vivo evaluation of a novel format of a bivalent HER3-targeting and albumin-binding therapeutic affibody construct. Sci Rep 2017; 7:43118. [PMID: 28230065 PMCID: PMC5322329 DOI: 10.1038/srep43118] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/19/2017] [Indexed: 01/21/2023] Open
Abstract
Overexpression of human epidermal growth factor receptor 3 (HER3) is involved in resistance to several therapies for malignant tumours. Currently, several anti-HER3 monoclonal antibodies are under clinical development. We introduce an alternative approach to HER3-targeted therapy based on engineered scaffold proteins, i.e. affibody molecules. We designed a small construct (22.5 kDa, denoted 3A3), consisting of two high-affinity anti-HER3 affibody molecules flanking an albumin-binding domain ABD, which was introduced for prolonged residence in circulation. In vitro, 3A3 efficiently inhibited growth of HER3-expressing BxPC-3 cells. Biodistribution in mice was measured using 3A3 that was site-specifically labelled with 111In via a DOTA chelator. The residence time of 111In-DOTA-3A3 in blood was extended when compared with the monomeric affibody molecule. 111In-DOTA-3A3 accumulated specifically in HER3-expressing BxPC-3 xenografts in mice. However, 111In-DOTA-3A3 cleared more rapidly from blood than a size-matched control construct 111In-DOTA-TAT, most likely due to sequestering of 3A3 by mErbB3, the murine counterpart of HER3. Repeated dosing and increase of injected protein dose decreased uptake of 111In-DOTA-3A3 in mErbB3-expressing tissues. Encouragingly, growth of BxPC-3 xenografts in mice was delayed in an experimental (pilot-scale) therapy study using 3A3. We conclude that the 3A3 affibody format seems promising for treatment of HER3-overexpressing tumours.
Collapse
|
93
|
Klement M, Zheng J, Liu C, Tan HL, Wong VVT, Choo ABH, Lee DY, Ow DSW. Antibody engineering of a cytotoxic monoclonal antibody 84 against human embryonic stem cells: Investigating the effects of multivalency on cytotoxicity. J Biotechnol 2017; 243:29-37. [PMID: 28042013 DOI: 10.1016/j.jbiotec.2016.12.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 11/22/2016] [Accepted: 12/27/2016] [Indexed: 02/06/2023]
Abstract
Antibody fragments have shown targeted specificity to their antigens, but only modest tissue retention times in vivo and in vitro. Multimerization has been used as a protein engineering tool to increase the number of binding units and thereby enhance the efficacy and retention time of antibody fragments. In this work, we explored the effects of valency using a series of self-assembling polypeptides based on the GCN4 leucine zipper multimerization domain fused to a single-chain variable fragment via an antibody upper hinge sequence. Four engineered antibody fragments with a valency from one to four antigen-binding units of a cytotoxic monoclonal antibody 84 against human embryonic stem cells (hESC) were constructed. We hypothesized that higher cytotoxicity would be observed for fragments with increased valency. Flow cytometry analysis revealed that the trimeric and tetrameric engineered antibody fragments resulted in the highest degree of cytotoxicity to the undifferentiated hESC, while the engineered antibody fragments were observed to have improved tissue penetration into cell clusters. Thus, a trade off was made for the trimeric versus tetrameric fragment due to improved tissue penetration. These results have direct implications for antibody-mediated removal of undifferentiated hESC during regenerative medicine and cell therapy.
Collapse
Affiliation(s)
- Maximilian Klement
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), 20 Biopolis Way, #06-01 Centros, 138668, Singapore; Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117576, Singapore
| | - Jiyun Zheng
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), 20 Biopolis Way, #06-01 Centros, 138668, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, 28 Medical Drive, #05-01, 117456, Singapore
| | - Chengcheng Liu
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), 20 Biopolis Way, #06-01 Centros, 138668, Singapore
| | - Heng-Liang Tan
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), 20 Biopolis Way, #06-01 Centros, 138668, Singapore
| | - Victor Vai Tak Wong
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), 20 Biopolis Way, #06-01 Centros, 138668, Singapore
| | - Andre Boon-Hwa Choo
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), 20 Biopolis Way, #06-01 Centros, 138668, Singapore; Department of Biomedical Engineering, National University of Singapore, 9 Engineering Drive 1, 117575, Singapore
| | - Dong-Yup Lee
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), 20 Biopolis Way, #06-01 Centros, 138668, Singapore; Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117576, Singapore; NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, 28 Medical Drive, 117456, Singapore.
| | - Dave Siak-Wei Ow
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), 20 Biopolis Way, #06-01 Centros, 138668, Singapore.
| |
Collapse
|
94
|
Lomonosova AV, Ulitin AB, Kazakov AS, Mirzabekov TA, Permyakov EA, Permyakov SE. Derivative of Extremophilic 50S Ribosomal Protein L35Ae as an Alternative Protein Scaffold. PLoS One 2017; 12:e0170349. [PMID: 28103321 PMCID: PMC5245882 DOI: 10.1371/journal.pone.0170349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/03/2017] [Indexed: 12/01/2022] Open
Abstract
Small antibody mimetics, or alternative binding proteins (ABPs), extend and complement antibody functionality with numerous applications in research, diagnostics and therapeutics. Given the superiority of ABPs, the last two decades have witnessed development of dozens of alternative protein scaffolds (APSs) for the design of ABPs. Proteins from extremophiles with their high structural stability are especially favorable for APS design. Here, a 10X mutant of the 50S ribosomal protein L35Ae from hyperthermophilic archaea Pyrococcus horikoshii has been probed as an APS. A phage display library of L35Ae 10X was generated by randomization of its three CDR-like loop regions (repertoire size of 2×108). Two L35Ae 10X variants specific to a model target, the hen egg-white lysozyme (HEL), were isolated from the resulting library using phage display. The affinity of these variants (L4 and L7) to HEL ranges from 0.10 μM to 1.6 μM, according to surface plasmon resonance data. While L4 has 1-2 orders of magnitude lower affinity to HEL homologue, bovine α-lactalbumin (BLA), L7 is equally specific to HEL and BLA. The reference L35Ae 10X is non-specific to both HEL and BLA. L4 and L7 are more resistant to denaturation by guanidine hydrochloride compared to the reference L35Ae 10X (mid-transition concentration is higher by 0.1-0.5 M). Chemical crosslinking experiments reveal an increased propensity of L4 and L7 to multimerization. Overall, the CDR-like loop regions of L35Ae 10X represent a proper interface for generation of functional ABPs. Hence, L35Ae is shown to extend the growing family of protein scaffolds dedicated to the design of novel binding proteins.
Collapse
Affiliation(s)
- Anna V. Lomonosova
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, Moscow region, Russia
| | | | - Alexei S. Kazakov
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, Moscow region, Russia
| | - Tajib A. Mirzabekov
- Antherix, Pushchino, Moscow region, Russia
- Biomirex Inc., Watertown, Massachusetts, United States of America
| | - Eugene A. Permyakov
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, Moscow region, Russia
| | - Sergei E. Permyakov
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, Moscow region, Russia
| |
Collapse
|
95
|
Abstract
Synthetic protein switches with tailored response functions are finding increasing applications as tools in basic research and biotechnology. With a number of successful design strategies emerging, the construction of synthetic protein switches still frequently necessitates an integrated approach that combines detailed biochemical and biophysical characterization in combination with high-throughput screening to construct tailored synthetic protein switches. This is increasingly complemented by computational strategies that aim to reduce the need for costly empirical optimization and thus facilitate the protein design process. Successful computational design approaches range from analyzing phylogenetic data to infer useful structural, biophysical, and biochemical information to modeling the structure and function of proteins ab initio. The following chapter provides an overview over the theoretical considerations and experimental approaches that have been successful applied in the construction of synthetic protein switches.
Collapse
Affiliation(s)
- Viktor Stein
- Fachbereich Biologie, Technische Universität Darmstadt, 64287, Darmstadt, Germany.
| |
Collapse
|
96
|
Teplyakov A, Malia TJ, Obmolova G, Jacobs SA, O'Neil KT, Gilliland GL. Conformational flexibility of an anti-IL-13 DARPin†. Protein Eng Des Sel 2016; 30:31-37. [PMID: 27881684 DOI: 10.1093/protein/gzw059] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 09/24/2016] [Accepted: 10/20/2016] [Indexed: 01/29/2023] Open
Abstract
Designed ankyrin repeat proteins (DARPin®) are artificial non-immunoglobulin binding proteins with potential applications as therapeutic molecules. DARPin 6G9 binds interleukin-13 with high affinity and blocks the signaling pathway and as such is promising for the treatment of asthma and other atopic diseases. The crystal structures of DARPin 6G9 in the unbound form and in complex with IL-13 were determined at high resolution. The DARPin competes for the same epitope as the IL-13 receptor chain 13Rα1 but does not interfere with the binding of the other receptor chain, IL-4Rα. Analysis of multiple copies of the DARPin molecule in the crystal indicates the conformational instability in the N-terminal cap that was predicted from molecular dynamics simulations. Comparison of the DARPin structures in the free state and in complex with IL-13 reveals a concerted movement of the ankyrin repeats upon binding resulted in the opening of the binding site. The induced-fit mode of binding employed by DARPin 6G9 is very unusual for DARPins since they were designed as particularly stable and rigid molecules. This finding shows that DARPins can operate by various binding mechanisms and suggests that some flexibility in the scaffold may be an advantage.
Collapse
Affiliation(s)
- Alexey Teplyakov
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477, USA
| | - Thomas J Malia
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477, USA
| | - Galina Obmolova
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477, USA
| | - Steven A Jacobs
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477, USA
| | - Karyn T O'Neil
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477, USA
| | - Gary L Gilliland
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477, USA
| |
Collapse
|
97
|
Zhang W, Ben-David M, Sidhu SS. Engineering cell signaling modulators from native protein-protein interactions. Curr Opin Struct Biol 2016; 45:25-35. [PMID: 27866084 DOI: 10.1016/j.sbi.2016.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 11/02/2016] [Indexed: 10/20/2022]
Abstract
Recent studies on genome sequencing and genetic screens with RNAi and CRISPR technology have revolutionized our understanding of aberrant signaling networks in human diseases. A strategy combining both genetic and protein-based technologies should be at the heart of modern drug-development efforts, particularly in the era of precision medicine. Thus, there is an urgent need for efficient platforms to develop probes that can modulate protein function in cells to validate drug targets and to develop therapeutic leads. Advanced protein engineering has enabled the rapid production of monoclonal antibodies and small protein scaffold affinity reagents for diverse protein targets. Here, we review the most recent progress on engineering natural protein-protein interactions for modulation of cell signaling.
Collapse
Affiliation(s)
- Wei Zhang
- The Donnelly Centre for Cellular and Biomolecular Research, Banting and Best Department of Medical Research, and Department of Molecular Genetics, University of Toronto, 160 College Street, Toronto, Ontario, M5S3E1, Canada
| | - Moshe Ben-David
- The Donnelly Centre for Cellular and Biomolecular Research, Banting and Best Department of Medical Research, and Department of Molecular Genetics, University of Toronto, 160 College Street, Toronto, Ontario, M5S3E1, Canada
| | - Sachdev S Sidhu
- The Donnelly Centre for Cellular and Biomolecular Research, Banting and Best Department of Medical Research, and Department of Molecular Genetics, University of Toronto, 160 College Street, Toronto, Ontario, M5S3E1, Canada.
| |
Collapse
|
98
|
Bourseau-Guilmain E, Menard JA, Belting M. Targeting of non-cycling cells of hypoxic tumors. Cell Cycle 2016; 15:2848-2849. [PMID: 27356029 DOI: 10.1080/15384101.2016.1204867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- E Bourseau-Guilmain
- a Department of Clinical Sciences , Section of Oncology and Pathology, Lund University , Lund , Sweden
| | - J A Menard
- a Department of Clinical Sciences , Section of Oncology and Pathology, Lund University , Lund , Sweden
| | - M Belting
- a Department of Clinical Sciences , Section of Oncology and Pathology, Lund University , Lund , Sweden.,b Department of Oncology and Radiotherapy , Skåne University Hospital , Lund , Sweden
| |
Collapse
|
99
|
Dias AM, Roque AC. The future of protein scaffolds as affinity reagents for purification. Biotechnol Bioeng 2016; 114:481-491. [DOI: 10.1002/bit.26090] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/25/2016] [Accepted: 08/26/2016] [Indexed: 01/07/2023]
Affiliation(s)
- Ana M.G.C. Dias
- UCIBIO, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia; Universidade Nova de Lisboa; Campus Caparica Caparica 2829-516 Portugal
| | - Ana C.A. Roque
- UCIBIO, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia; Universidade Nova de Lisboa; Campus Caparica Caparica 2829-516 Portugal
| |
Collapse
|
100
|
Zadravec P, Marečková L, Petroková H, Hodnik V, Perišić Nanut M, Anderluh G, Štrukelj B, Malý P, Berlec A. Development of Recombinant Lactococcus lactis Displaying Albumin-Binding Domain Variants against Shiga Toxin 1 B Subunit. PLoS One 2016; 11:e0162625. [PMID: 27606705 PMCID: PMC5015993 DOI: 10.1371/journal.pone.0162625] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/25/2016] [Indexed: 01/06/2023] Open
Abstract
Infections with shiga toxin-producing bacteria, like enterohemorrhagic Escherichia coli and Shigella dysenteriae, represent a serious medical problem. No specific and effective treatment is available for patients with these infections, creating a need for the development of new therapies. Recombinant lactic acid bacterium Lactococcus lactis was engineered to bind Shiga toxin by displaying novel designed albumin binding domains (ABD) against Shiga toxin 1 B subunit (Stx1B) on their surface. Functional recombinant Stx1B was produced in Escherichia coli and used as a target for selection of 17 different ABD variants (named S1B) from the ABD scaffold-derived high-complex combinatorial library in combination with a five-round ribosome display. Two most promising S1Bs (S1B22 and S1B26) were characterized into more details by ELISA, surface plasmon resonance and microscale thermophoresis. Addition of S1Bs changed the subcellular distribution of Stx1B, completely eliminating it from Golgi apparatus most likely by interfering with its retrograde transport. All ABD variants were successfully displayed on the surface of L. lactis by fusing to the Usp45 secretion signal and to the peptidoglycan-binding C terminus of AcmA. Binding of Stx1B by engineered lactococcal cells was confirmed using flow cytometry and whole cell ELISA. Lactic acid bacteria prepared in this study are potentially useful for the removal of Shiga toxin from human intestine.
Collapse
Affiliation(s)
- Petra Zadravec
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, SI-1000, Ljubljana, Slovenia
- The Chair of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, SI-1000, Ljubljana, Slovenia
| | - Lucie Marečková
- Laboratory of Ligand Engineering, Institute of Biotechnology CAS, v. v. i., BIOCEV Research Center, Průmyslová 595, 252 50, Vestec, Czech Republic
| | - Hana Petroková
- Laboratory of Ligand Engineering, Institute of Biotechnology CAS, v. v. i., BIOCEV Research Center, Průmyslová 595, 252 50, Vestec, Czech Republic
| | - Vesna Hodnik
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
- National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Milica Perišić Nanut
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Gregor Anderluh
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
- National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Borut Štrukelj
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, SI-1000, Ljubljana, Slovenia
- The Chair of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, SI-1000, Ljubljana, Slovenia
| | - Petr Malý
- Laboratory of Ligand Engineering, Institute of Biotechnology CAS, v. v. i., BIOCEV Research Center, Průmyslová 595, 252 50, Vestec, Czech Republic
| | - Aleš Berlec
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, SI-1000, Ljubljana, Slovenia
- * E-mail:
| |
Collapse
|