14 research outputs found

    Using protein geometry to optimize cytotoxicity and the cytokine window of a ROR1 specific T cell engager

    Get PDF
    T cell engaging bispecific antibodies have shown clinical proof of concept for hematologic malignancies. Still, cytokine release syndrome, neurotoxicity, and on-target-off-tumor toxicity, especially in the solid tumor setting, represent major obstacles. Second generation TCEs have been described that decouple cytotoxicity from cytokine release by reducing the apparent binding affinity for CD3 and/or the TAA but the results of such engineering have generally led only to reduced maximum induction of cytokine release and often at the expense of maximum cytotoxicity. Using ROR1 as our model TAA and highly modular camelid nanobodies, we describe the engineering of a next generation decoupled TCE that incorporates a “cytokine window” defined as a dose range in which maximal killing is reached but cytokine release may be modulated from very low for safety to nearly that induced by first generation TCEs. This latter attribute supports pro-inflammatory anti-tumor activity including bystander killing and can potentially be used by clinicians to safely titrate patient dose to that which mediates maximum efficacy that is postulated as greater than that possible using standard second generation approaches. We used a combined method of optimizing TCE mediated synaptic distance and apparent affinity tuning of the TAA binding arms to generate a relatively long but persistent synapse that supports a wide cytokine window, potent killing and a reduced propensity towards immune exhaustion. Importantly, this next generation TCE induced significant tumor growth inhibition in vivo but unlike a first-generation non-decoupled benchmark TCE that induced lethal CRS, no signs of adverse events were observed

    Untersuchungen zur Substrattoleranz von Sactisynthasen für die Generierung Thioether‐verbrückter Sactipeptide mit neuen Eigenschaften

    Get PDF
    Sactipeptide sind ribosomal synthetisierte Peptide, die eine einzigartige Verknüpfung von Schwefel und α‐Kohlenstoffen enthalten. Die Bildung von Thioetherbrücken wird in diesen Molekülen durch Sactisynthasen katalysiert. Diese spezielle Art der Verknüpfung verleiht Sactipeptiden eine erhöhte strukturelle, thermische und proteolytische Stabilität, was sie zu attraktiven Gerüsten für die Entwicklung neuer Biotherapeutika macht. In diesem Artikel berichten wir über eine Studie zur Substrattoleranz der Sactisynthase AlbA, die die Bildung von Thioetherbrücken im Sactipeptid Subtilosin A katalysiert. Wir haben eine Modifikationsstelle innerhalb dieses Sactipeptids identifiziert, die ein Peptid‐Engineering ohne Beeinträchtigung der Bildung von Thioetherbrücken ermöglicht. Eine Reihe von natürlichen und hybriden Sactipeptidkonstrukten wurde hergestellt, um die AlbA vermittelte Bildung von Thioetherbrücken zu untersuchen und diese massenspektrometrisch zu identifizieren. In einer Proof‐of‐Principle‐Studie haben wir Subtilosin A mit einer neue Funktion ausgestattet, ein Thioether‐verbrücktes Streptavidin‐bindendes Peptid generiert und damit die Tür für das funktionelle Engineering von Sactipeptiden weiter geöffnet

    Sactipeptide Engineering by Probing the Substrate Tolerance of a Thioether‐Bond‐Forming Sactisynthase

    Get PDF
    Sactipeptides are ribosomally synthesized peptides containing a unique sulfur to α‐carbon crosslink. Catalyzed by sactisynthases, this thioether pattern endows sactipeptides with enhanced structural, thermal, and proteolytic stability, which makes them attractive scaffolds for the development of novel biotherapeutics. Herein, we report the in‐depth study on the substrate tolerance of the sactisynthase AlbA to catalyze the formation of thioether bridges in sactipeptides. We identified a possible modification site within the sactipeptide subtilosin A allowing for peptide engineering without compromising formation of thioether bridges. A panel of natural and hybrid sactipeptides was produced to study the AlbA‐mediated formation of thioether bridges, which were identified mass‐spectrometrically. In a proof‐of‐principle study, we re‐engineered subtilosin A to a thioether‐bridged, specific streptavidin targeting peptide, opening the door for the functional engineering of sactipeptides

    Self-decorating cells via surface-initiated enzymatic controlled radical polymerization

    Get PDF
    Through the innovative use of surface-displayed horseradish peroxidase, this work explores the enzymatic catalysis of both bioRAFT polymerization and bioATRP to prompt polymer synthesis on the surface of Saccharomyces cerevisiae cells, with bioATRP outperforming bioRAFT polymerization. The resulting surface modification of living yeast cells with synthetic polymers allows for a significant change in yeast phenotype, including growth profile, aggregation characteristics, and conjugation of non-native enzymes to the clickable polymers on the cell surface, opening new avenues in bioorthogonal cell-surface engineering

    Influence of wastewater discharge on the occurrence of PBTC, HEDP, and aminophosphonates in sediment, suspended matter, and the aqueous phase of rivers

    No full text
    Sediment, suspended matter (SM), and water of a large river (Neckar; River1) and a small river (Körsch; River2) were analyzed for the phosphonates 2-phosphonobutane-1,2,4-tricarboxylic acid (PBTC), 1-hydroxyethylidene (1,1-diphosphonic acid) (HEDP), aminotris (methylphosphonic acid) (ATMP), ethylenediaminetetra (methylene phosphonic acid) (EDTMP), and diethylenetriaminepenta (methylene phosphonic acid) (DTPMP). Ten samplings were performed at intervals of one to two months during one year, each covering the relevant matrices before and behind the discharge point of a wastewater treatment plant (WWTP). In River1, the total concentration of dissolved phosphonate did not change significantly (2.4–5.8 µg/L before vs. 2.5–6.6 µg/L behind WWTP; p = 0.9360). In River2, it increased significantly from 2000 mg/kg phosphonate loads. In general, the nitrogen-free phosphonates PBTC and HEDP were most predominant in both dissolved and adsorbed form, of which HEDP had the highest adsorption affinity

    A tightly regulated and adjustable CRISPR-dCas9 based AND gate in yeast

    Get PDF
    The robust and precise on and off switching of one or more genes of interest, followed by expression or repression is essential for many biological circuits as well as for industrial applications. However, many regulated systems published to date influence the viability of the host cell, show high basal expression or enable only the overexpression of the target gene without the possibility of fine regulation. Herein, we describe an AND gate designed to overcome these limitations by combining the advantages of three well established systems, namely the scaffold RNA CRISPR/dCas9 platform that is controlled by Gal10 as a natural and by LexA-ER-AD as heterologous transcription factor. We hence developed a predictable and modular, versatile expression control system. The selection of a reporter gene set up combining a gene of interest (GOI) with a fluorophore by the ribosomal skipping T2A sequence allows to adapt the system to any gene of interest without losing reporter function. In order to obtain a better understanding of the underlying principles and the functioning of our system, we backed our experimental findings with the development of a mathematical model and single-cell analysis

    DataSheet_1_Using protein geometry to optimize cytotoxicity and the cytokine window of a ROR1 specific T cell engager.pdf

    No full text
    T cell engaging bispecific antibodies have shown clinical proof of concept for hematologic malignancies. Still, cytokine release syndrome, neurotoxicity, and on-target-off-tumor toxicity, especially in the solid tumor setting, represent major obstacles. Second generation TCEs have been described that decouple cytotoxicity from cytokine release by reducing the apparent binding affinity for CD3 and/or the TAA but the results of such engineering have generally led only to reduced maximum induction of cytokine release and often at the expense of maximum cytotoxicity. Using ROR1 as our model TAA and highly modular camelid nanobodies, we describe the engineering of a next generation decoupled TCE that incorporates a “cytokine window” defined as a dose range in which maximal killing is reached but cytokine release may be modulated from very low for safety to nearly that induced by first generation TCEs. This latter attribute supports pro-inflammatory anti-tumor activity including bystander killing and can potentially be used by clinicians to safely titrate patient dose to that which mediates maximum efficacy that is postulated as greater than that possible using standard second generation approaches. We used a combined method of optimizing TCE mediated synaptic distance and apparent affinity tuning of the TAA binding arms to generate a relatively long but persistent synapse that supports a wide cytokine window, potent killing and a reduced propensity towards immune exhaustion. Importantly, this next generation TCE induced significant tumor growth inhibition in vivo but unlike a first-generation non-decoupled benchmark TCE that induced lethal CRS, no signs of adverse events were observed.</p

    Table_1_Using protein geometry to optimize cytotoxicity and the cytokine window of a ROR1 specific T cell engager.xlsx

    No full text
    T cell engaging bispecific antibodies have shown clinical proof of concept for hematologic malignancies. Still, cytokine release syndrome, neurotoxicity, and on-target-off-tumor toxicity, especially in the solid tumor setting, represent major obstacles. Second generation TCEs have been described that decouple cytotoxicity from cytokine release by reducing the apparent binding affinity for CD3 and/or the TAA but the results of such engineering have generally led only to reduced maximum induction of cytokine release and often at the expense of maximum cytotoxicity. Using ROR1 as our model TAA and highly modular camelid nanobodies, we describe the engineering of a next generation decoupled TCE that incorporates a “cytokine window” defined as a dose range in which maximal killing is reached but cytokine release may be modulated from very low for safety to nearly that induced by first generation TCEs. This latter attribute supports pro-inflammatory anti-tumor activity including bystander killing and can potentially be used by clinicians to safely titrate patient dose to that which mediates maximum efficacy that is postulated as greater than that possible using standard second generation approaches. We used a combined method of optimizing TCE mediated synaptic distance and apparent affinity tuning of the TAA binding arms to generate a relatively long but persistent synapse that supports a wide cytokine window, potent killing and a reduced propensity towards immune exhaustion. Importantly, this next generation TCE induced significant tumor growth inhibition in vivo but unlike a first-generation non-decoupled benchmark TCE that induced lethal CRS, no signs of adverse events were observed.</p

    Synthesis of artificial cells via biocatalytic polymerisation-induced self-assembly

    No full text
    Artificial cells are biomimetic microstructures that mimic functions of natural cells and find application, e.g., as microreactors, as building blocks for molecular systems engineering, and to host synthetic biology pathways. Here, we report enzymatically synthesised polymer-based artificial cells with the ability to express proteins. They are created by biocatalytic atom transfer radical polymerization-induced self-assembly (bioPISA). The metalloprotein myoglobin synthesises amphiphilic block copolymers that self-assemble into structures ranging from micelles over worm-like micelles to polymersomes and giant unilamellar vesicles (GUVs). The GUVs encapsulate cargo during the polymerisation, including enzymes, nanoparticles, microparticles, plasmids and cell lysate. The resulting artificial cells act as microreactors for enzymatic reactions and for osteoblast-inspired biomineralization, and could express proteins when fed with amino acids, as shown by the expression of the fluorescent protein mClover and of actin. Actin polymerises in the vesicles and alters the artificial cell’s internal structure by creating internal compartments. Thus, bioPISA-derived GUVs mimic bacteria as they are composed of a microscopic reaction compartment that contains genetic information which is able to express proteins upon induction. bioPISA not only is a powerful tool in the pursuit of artificial cells but also for the easy and highly efficient encapsulation of biological molecules under mild conditions and in biologically relevant media. Therefore, it could have significant implications for the development of biomaterials and drug-delivery systems, as well as for cell encapsulation, and the in-situ formation of nano-objects

    Artificial cell synthesis using biocatalytic polymerization-induced self-assembly

    No full text
    Artificial cells are biomimetic microstructures that mimic functions of natural cells, can be applied as building blocks for molecular systems engineering, and host synthetic biology pathways. Here we report enzymatically synthesized polymer-based artificial cells with the ability to express proteins. Artificial cells were synthesized using biocatalytic atom transfer radical polymerization-induced self-assembly, in which myoglobin synthesizes amphiphilic block co-polymers that self-assemble into structures such as micelles, worm-like micelles, polymersomes and giant unilamellar vesicles (GUVs). The GUVs encapsulate cargo during the polymerization, including enzymes, nanoparticles, microparticles, plasmids and cell lysate. The resulting artificial cells act as microreactors for enzymatic reactions and for osteoblast-inspired biomineralization. Moreover, they can express proteins such as a fluorescent protein and actin when fed with amino acids. Actin polymerizes in the vesicles and alters the artificial cells’ internal structure by creating internal compartments. Thus, biocatalytic atom transfer radical polymerization-induced self-assembly-derived GUVs can mimic bacteria as they are composed of a microscopic reaction compartment that contains genetic information for protein expression upon induction
    corecore