www.slas2020.org Open in urlscan Pro
69.63.145.61  Public Scan

URL: https://www.slas2020.org/program/slas2020-scientific-podium-program/podium-abstract-compendium/
Submission: On November 21 via manual from US — Scanned from DE

Form analysis 1 forms found in the DOM

POST /site-search/?nocache=1#searchForm

<form id="searchForm" method="post" class="pform navbar-form " role="search" action="/site-search/?nocache=1#searchForm">
  <div class="form-group">
    <div class="input-group">
      <input type="text" name="Keywords" id="txtKeywords" class="form-control" value="" placeholder="Search">
      <span class="input-group-btn">
        <button type="submit" class="btn btn-default"> Go! </button>
      </span>
    </div>
  </div>
</form>

Text Content

 

SLAS HOME

Go!
 * Conference App
 * Event Scheduler
   

Toggle navigation
 * Conference
   * Photo Gallery
   * SLAS2020 Code of Conduct
   * Schedule-at-a-Glance
   * What's New for 2020
   * SLAS2020 Conference App
   * For Students and Early Career Professionals
   * Tuesday Evening Celebration by Hamilton
   * Scientific Program Committee
 * Program
   * ePoster Gallery
   * Educational Program
   * Event Scheduler
   * Scientific Podium Program
     * Podium Abstract Compendium
   * Short Course Program
   * SLAS Ignite
     * SLAS Ignite Panel Discussions
     * SLAS Ignite Partnership Program
   * Special Interest Groups
 * Exhibition
   * SLAS2020 Exhibition
   * Exhibition Map & Participating Companies
   * New Product Announcements at SLAS2020
   * Exhibitor Tutorial Program
   * Solutions Spotlight Presentations
 * For Exhibitors
   * General Information
   * Innovation AveNEW for Startups
     * Innovation AveNEW Participating Companies
 * Sponsors
   * Sponsorship Prospectus
   * 2020 Sponsors
 * Awards
   * General Information
 * Contact Us
   * Contact Us

 * Conference App
 * Event Scheduler
   




PODIUM ABSTRACT COMPENDIUM

Advances in Bioanalytics and Biomarkers Assay Development and Screening
Automation and High Throughput Technologies Biologics Discovery Cellular
Technologies Data Analysis and Informatics Drug Target Strategies Micro- and
Nano Technologies Molecular Libraries Precision Medicine Technologies

Podium presentations are organized into 10 educational tracks. Podium abstracts
and speaker information are organized first by track and then by session below.

To search for a specific speaker use the 'Find' functionality in your browser
(usually Ctrl + F).

To view a complete schedule of podium presentations and schedule of events for
SLAS2020 and to view speaker bios and photos, please visit the SLAS2020 Event
Scheduler.


ADVANCES IN BIOANALYTICS AND BIOMARKERS

Track Chair(s): Andreas Luippold, Ph.D., Boehringer Ingelheim (Germany) and
Martin Giera, Ph.D., Leiden University Medical Center (The Netherlands)

LABEL-FREE BIOANALYTICAL TECHNIQUES

Session Chair: Daniel Bischoff, Ph.D., Boehringer Ingelheim (Germany)

 * Combining Arrays and Mass Spectrometry for High-Throughput Experiments in
   Chemistry and Biology
   Milan Mrksich, Northwestern University
   
   This talk will describe an approach for using mass spectrometry and arrays of
   self-assembled monolayers to perform quantitative experiments in
   high-throughput. The arrays are prepared by immobilizing small molecules,
   proteins, peptides and carbohydrates to self-assembled monolayers of
   alkanethiols on gold. These arrays are then treated with reactants—either
   chemical reagents or enzymes—and then analyzed using the SAMDI technique to
   identify the masses of substituted alkanethiolates in the monolayer and
   therefore a broad range of reactivities and post-translational
   modifications—including kinase, protease, methyltransferase and
   carbohydrate-directed modifications—and for discovering chemical reactions.
   This talk will describe applications to high throughput experiments,
   including the discovery of reactions, the use of carbohydrate arrays to
   discover novel enzymes, the preparation of peptide arrays to profile the
   enzyme activities in cell lysates and high-throughput screening to discover
   novel reactions and small molecular modulators.
   
   These examples illustrate the broad capability of the SAMDI method to profile
   and discover molecular activities in the molecular sciences.

 * Establishing Capabilities for (Ultra)High-Throughput Mass Spectrometry at
   Merck: Reflections from “Year One”
   David McLaren, MSD.
   
   Long a prized analytical tool for low and medium-throughput applications,
   recent innovations in mass spectrometry (MS) are changing the way
   pharmaceutical scientists apply MS to challenges requiring truly
   high-throughput. Such endeavors include HTS, parallel medicinal chemistry and
   protein engineering with emerging needs in drug metabolism and
   pharmacokinetics as well. At Merck, we have embarked on a journey to
   incorporate these innovative uHT-MS capabilities in our own approach to drug
   discovery. This presentation will highlight how we have deployed both a
   fully-automated uHT-MALDI-MS instrument and the Echo-MS platform in a
   cross-functional research setting.
   
   As one example, we will demonstrate the application of the uHT-MALDI-MS to a
   prototypical HTS assay for a kinase target. Here we will discuss how we have
   enabled "on-demand" MALDI target plate preparation in both 384w and 1536w
   formats and will further compare & contrast this data to those obtained using
   a conventional Glo assay for both primary (single concentration) screening
   and compound titrations.
   
   We will also describe how we have leveraged the Echo-MS platform to triage
   hits from an HTS designed to discover inhibitors of lipid metabolism which
   employed a fluorescent, cell-free assay. Here, we will highlight a useful
   “quench-and-read” sample preparation approach that allows for the selective
   enrichment of hydrophobic analytes and which is also fully compatible with
   direct acoustic transfer of the surface solution to the mass spectrometer for
   analysis.
   
   Throughout the presentation, we will highlight the relevant figures of merit
   for each application and discuss our findings around the ‘critical
   parameters’ whose optimization was important to enabling assay
   success. Finally, we will conclude with a perspective of the future of our
   journey in this space.

 * CETSA® Beyond Soluble Targets: A Broad Application to Multi-Pass
   Transmembrane Proteins
   Aarti Kawatkar, AstraZeneca
   
   Demonstration of target binding is a key requirement for understanding the
   mode of action of new therapeutics. The cellular thermal shift assay (CETSA®)
   has been introduced as a powerful label-free method to assess target
   engagement in physiological environments. Here, we present the application of
   live-cell CETSA® to different classes of integral multi-pass transmembrane
   proteins using three case studies: the first showing a large and robust
   stabilization of the outer mitochondrial five-pass transmembrane protein
   TSPO, the second being a modest stabilization of SERCA2 and the last
   describing an atypical compound-driven stabilization of the GPCR PAR2. Our
   data demonstrated that using modified protocols with detergent extraction
   after the heating step, CETSA® can reliably be applied to several membrane
   proteins of different complexity. By showing examples with distinct CETSA®
   behaviors, we aim to provide the scientific community with an overview of
   different scenarios to expect during CETSA® experiments, especially for
   challenging, membrane-bound targets.

 * Novel Label-Free Interaction Technologies and Concepts and its Direct Impact
   on Early Drug Discovery
   Anders Gunnarsson, AstraZeneca
   
   This presentation will disclose three new biophysical concepts and
   technologies currently implemented in early drug discovery at AstraZeneca to
   significantly enhance throughput, broaden chemical space or provide an
   in-depth mechanistic understanding of drug targets and its oligomeric state
   at the single-molecule level.
   
   * Profiling >100 compounds at the speed and cost of 1.
     Concentration-independent off-rate screening using optical biosensors
     enable small scale (µmol) library synthesis followed by kinetic profiling
     of the crude reaction products to rapidly inform structure-activity
     relations (SAR) within a day of synthesis start.
   * Overcoming limitations of profiling potent or covalent compounds with
     surface-based biosensors. Using novel streptavidin-based concepts for
     regeneratable surface chemistry significantly enhance throughput for potent
     (long residence time) compounds and even enable assessment of covalent
     compounds.
   * Weighing single proteins by light. Label-free single-molecule scattering
     measurements provide rapid information on the oligomeric state of proteins
     under biochemical assay conditions and provide instant information on small
     molecule-induced complex formation of unlabeled proteins.

TARGET ENGAGEMENT AND PATHWAYS 

Session Chair: Gary Siuzdak, Ph.D., Scripps Research (USA)

 * Activity Metabolomics
   Gary Siuzdak, Scripps Research
   
   The metabolome, the small molecule chemical entities involved in metabolism,
   has traditionally been studied to identify biomarkers in the diagnosis and
   prediction of disease. However, the value of metabolomics has been redefined
   from a simple biomarker identification tool to a technology for the discovery
   of active drivers of biological processes. In this presentation, I will
   describe the molecular mechanisms by which the active cell metabolome affects
   cellular physiology through modulation of other "omic" levels, including the
   genome, epigenome, transcriptome and proteome. This concept of activity
   screening guided by metabolomics to identify biologically active metabolites
   or “activity metabolomics”, is having a broad impact on biology.

 * Connecting High-Throughput Screening and Clinical Pharmacology Using Stable
   Isotope Tracer Kinetics and Mass Spectrometry: Moving from Enzyme Activity to
   in vivo Pathway Flux with the Same Assay.
   Stephen Previs, MSD
   
   Stable isotope-labeled substrates can be of broad use in cases where
   target-based high-throughput screening aims to identify compounds that can
   modulate enzyme activity. For example, depending on the source of a given
   enzyme target, the presence of endogenous substrates or products can limit
   one’s ability to follow substrate product conversions; utilization of a
   labeled substrate(s) can help overcome background contamination. These same
   isotope flux assays can then be used to follow the progression of hits in
   later stages of development, including cell-based assays and in vivo studies.
   Although stable isotope tracer kinetics, coupled with mass spectrometry-based
   detection, can, therefore, connect all phases of drug discovery some caveats
   should be recognized to ensure reliable data interpretations.
   
   Our presentation will highlight key areas where the logic surrounding tracer
   kinetics diverges as the application of flux analyses moves across different
   stages of drug discovery. We will consider a case study that is focused on
   lipid biology, i.e. modulating the level of glycosylated ceramides. We will
   first outline how labeled substrates can be used to circumvent problems that
   arise in early screening. We will then outline how tracers can be used to
   progress molecules into later phases, including in vivo studies. Although one
   can use virtually the same back-end mass spectrometry assay to measure the
   formation of labeled products, several parameters change with regards to
   dosing the labeled substrates. For example, when measuring enzyme activity in
   early biochemical screening one needs to only measure the labeled product. In
   contrast, in vivo studies must contend with the fact that substantial amounts
   of “cold” (endogenous) substrate can exist, also, it may not be possible to
   maintain a steady-state exposure to the labeled substrate. Consequently,
   strategies need to account for temporal tracer dilution, most of which may
   not be immediately obvious and/or difficult to correct.
   
   In summary, the ability to measure stable isotope flux from precursors to
   products can provide a bridge that spans the entire spectrum of drug
   discovery and development. However, changes in the generation of a labeled
   product do not immediately reflect changes in the metabolic activity of a
   given target enzyme or pathway, it is possible to observe differences in the
   abundance of a labeled product which reflect an unexpected modulation of
   precursor metabolism. Although the example described here is focused on a
   targeted screen, the logic has immediate implications with regards to
   phenotypic screening; attention to a few details can influence essential
   decision points.

 * Novel Approaches to Quantitative Metabolomics
   Loren Olson, Sciex
   
   The major challenge in the field of metabolomics is to accurately identify
   and quantify hundreds of metabolites in a single run. Recently variable
   window SWATH acquisition has shown to identify a higher number of metabolites
   compared to the traditional Data Dependent Acquisition (DDA) approach, thus
   enabling broader metabolome coverage. Here we have implemented a variable
   window SWATH acquisition method for enhanced quantitation of selected
   metabolites using MS/MS, with reduced matrix interferences and improved
   signal-to-noise. Using MS/MS fragments for metabolite quantitation provides
   better selectivity, and ultimately increased sensitivity. Variable window
   SWATH Acquisition provided quality quantitative data for metabolites in
   complex matrix. Due to many coeluting metabolites in complex matrix, using
   only the MS spectrum and retention time is often not sufficient for
   metabolite identification. MS/MS information is necessary to obtain further
   structural knowledge about the metabolite. Complete full scan MS and MS/MS
   data is available in every SWATH file for improved ID. In addition, MS/MS
   quantitation of metabolites often leads to lower detection limits due to
   significantly improved signal to noise ratios vs MS data. Measuring the whole
   MS/MS spectrum allows selection of the best fragments for metabolite
   quantitation. SCIEX OS software combines comprehensive qualitative and
   quantitative data analysis, making data processing easier and more efficient.
   SWATH Acquisition on all detectable metabolites is successfully utilized for
   identification, and accurate MS/MS level quantification of metabolites in
   urine.

 * NMR-Based Metabolomics in Drug Research: Cancer Metabolism
   Martin Giera, Leiden University Medical Center
   
   Metabolism and in particular central energy metabolism have evolved as
   promising drug targets. A cutting edge technology that has become widely used
   for studying oxygen consumption and extracellular acidification is the
   Seahorse™ analyzer. While this technology allows for rapid label-free
   screening it does not provide further details on the involved metabolites and
   pathways. The quantitative analysis of these pathways, mainly involving
   glycolysis, the tricarboxylic acid cycle (TCA) and adjacent pathways is
   intrinsically very challenging. Several commercial solutions have evolved
   over the years predominantly using mass spectrometry and a series of labeled
   internal standards. However, many of these approaches suffer from long
   analytical procedures and the need for special internal standards or kits. As
   an alternative, we will discuss our NMR based workflow allowing the
   quantitative analysis of several important pathways for example glycolysis,
   TCA cycle, OxPhos, one-carbon metabolism and others. Our NMR based workflow
   allows for the rapid and quantitative analysis of >80 metabolites without the
   need for specialized kits or internal standards. The workflow can partially
   be operated in an automated fashion using a KNIME workflow such as KIMBLE.
   Moreover, flux analysis using 13C labeled materials can easily be adapted
   resulting in kinetic information.
   
   As an example for the usefulness of this workflow, we will discuss the
   discovery of choline kinase α (CHKA) as a possible target for the prevention
   of epithelial to mesenchymal transition (EMT) an important metastatic
   process. Using NMR based analysis of metabolic changes during TGFα induced
   EMT we could observe significant alterations in choline phosphorylation.
   Following up on these results by using experimental inhibitors we could
   identify CHKA as a crucial enzyme for the EMT phenotype.

TARGET AND MECHANISM IDENTIFICATION AFTER PHENOTYPIC SCREENS

Session Chair: Paul Tesar, Ph.D., Case Western Reserve University (USA)

 * Identifying Compounds that Improve Neuromuscular Function
   Lee Rubin, Dept of Stem Cell and Regenerative Biology, Harvard University;
   Harvard Stem Cell Institute
   
   Motor neuron diseases, as a class, are becoming increasingly well-known and
   understood. However, effective therapeutics that preserve motor neurons are
   still lacking. Motor neuron dysfunction is reflected, ultimately, in skeletal
   muscle weakness and deterioration. Surprisingly, the involvement of muscle
   cells themselves in diseases ranging from Spinal Muscular Atrophy, a
   childhood developmental disorder, to sarcopenia – muscle weakness with aging
   – is much less understood. I will describe two different projects – one
   directed at promoting the survival of healthy motor neurons, the other at
   improving muscle health. Both projects started with disease-relevant
   cell-based screens and culminated in the discovery of new therapeutic
   targets.

 * Can Lattice Theory Help Find a Cure for Paralysis?
   Nicola Richmond, GlaxoSmithKline
   
   With the advent of the Human Genome Project came the industrialization of the
   drug discovery process and a belief that combinatorial chemistry and high
   throughput screening would deliver molecules with increased potency against a
   single target of interest. Yet the attrition rate is still at the 10% mark
   and there remain many human diseases for which no effective treatment exists.
   As Swinney et. al. showed [1], there is compelling evidence that
   first-in-class drugs are more likely to be found by assays that measure a
   clinically meaningful phenotype in a physiologically relevant system rather
   than a single target-based screening approach in an artificial setting. One
   perceived issue with phenotypic screening is the lack of mechanistic
   knowledge. Whilst understanding mechanism of action (MOA) is not a
   prerequisite for FDA approval, it can guide a medicinal chemistry effort,
   predict potential toxicities and help define patient populations for clinical
   trials and ultimately the market place. There are several in vitro approaches
   to target deconvolution. However, these tend to be of lower throughput and
   better placed later in a screening cascade. So there is a real need for in
   silico-based approaches that can be deployed early on in a drug discovery
   program to identify potential MOAs. Using publicly available data on the
   Published Kinase Inhibitor Set (PKIS) [2,3], we describe the application of
   Formal Concept Analysis (FCA), an association mining technique with roots in
   set theory, to the problem of deconvoluting a phenotypic screen. We describe
   each compound in the PKIS by the set of kinases it inhibits. We then
   construct a Galois Lattice, whose nodes correspond to a set of compounds
   inhibiting a common set of kinases and where two nodes are connected if the
   compound set of the child node is a subset of the compound set of the parent
   node. Lattice nodes enriched with compounds that promote neurite outgrowth in
   rat inform which kinases should be targeted when seeking small molecules that
   encourage CNS axon repair following injury. The targets we identify using
   this push-button approach, that can be placed in the hands of the bench
   scientist, are in line with those identified in [3] and confirmed in siRNA
   studies.
   
   1. Swinney DC, Anthony J: How were new medicines discovered? Nat Rev Drug
   Discov 2011, 10(7):507-19.
   2. Drewry DH, Willson TM, Zuercher WJ: Seeding collaborations to advance
   kinase science with the GSK Published Kinase Inhibitor Set (PKIS). Curr Top
   Med Chem 2014, 14(3):340-2.
   3. Al-Ali H, Lee DH, Danzi MC, Nassif H, Gautam P, Wennerberg K, Zuercher WJ,
   Drewry DH, Lee JK, Lemmon VP, Bixby JL: Rational Polypharmacology:
   Systematically Identifying and Engaging Multiple Drug Targets To Promote Axon
   Growth. ACS Chem Biol 2015, 10(8): 1939-51.

 * Cytosolic Proteome & Affinity-Based Target Identification (CPATI)
   Xianshu Yang, MSD
   
   Disease-relevant phenotypic screening directly identifies ligands that
   modulate useful biology and constitutes a promising approach to the discovery
   of novel pharmaceutical treatments. Beyond identifying chemicals with
   desirable effects, it is important to identify the target(s) and mechanisms
   that drive the desirable phenotype in complex cellular systems. However,
   determining the relevant target(s) of phenotypically active ligands has often
   proven slow or impossible, hampering drug discovery and development.
   Recently, a few methodologies have emerged that enable detection of target
   engagement in cells, but most of them require prior chemical modification of
   either biologically active compounds or proteins. Here, we reported a novel
   cytosolic proteome & affinity-based target identification platform (CPATI),
   which is an unbiased, label-free and modification-free approach.
   
   We applied CPATI to identify the candidate target protein(s) of three
   compounds with Jurkat cells. First, native size-exclusion chromatography
   (SEC) was used to separate cellular cytosol isolated from Jurkat cell lysis.
   Second, using affinity-selection technology with our two-dimensional LC-MS
   system, three compounds were screened with 170 SEC fractions. Cytosolic
   fractions identified to have specific ligand binding were analyzed via
   quantitative proteomics. A combination of the protein-bound ligand recovery
   and target protein SEC elution profile revealed potential targets of test
   compounds. Third, the thermal shift experiment was conducted to identify
   proteins with elevated Tm in the presence of ligands, yielding a shortlist of
   target proteins. Fourth, the target proteins were selected for recombinant
   protein production and were validated in a binding assay with ligands.
   Finally, top target proteins were recommended for further validation.
   
   Three control ligands, compound A, compound B, and compound C were identified
   to bind specifically with Jurkat cytosolic fractions. Their associated target
   proteins NUDT1 (compound A), HSP90 (compound B), and PAK4 (compound C) were
   identified from the Jurkat cytosol as their top candidate targets,
   respectively. Compound A and compound B had a similar binding affinity (Kds)
   with specific cytosolic fractions and recombinant proteins NUDT1 and
   HSP90AA1/AB1, respectively. PPP3A-HSP90-CCT complexes were also identified.
   Compound C is an ATP competitive kinase inhibitor and was identified to
   associate with seventeen target proteins. Three recombinant proteins PRKACB,
   PRKCQ and STK38 were confirmed as compound C-bound target proteins.
   
   We also compared thermal shift experiments using compound C with compound
   C-bound cytosolic fractions and Jurkat crude cytosol. Former samples
   demonstrated advantage over later samples with short potential target
   proteins and less false positive. This technology potentially provides a
   broad application in target and biomarker identification from cells and
   tissues.

 * Combining Large-Scale in vitro Pharmacological Profiling and Human Cell-Based
   Phenotypic Profiling Identifies Novel Mechanisms of Cardiovascular Toxicity
   Ellen Berg, Eurofins Discovery
   
   We have previously described a phenotypic signature associated with
   cardiovascular toxicity relevant to vascular calcification and
   atherosclerosis from a human primary cell-based coronary artery smooth muscle
   cell model of vascular inflammation (BioMAP® CASM3C system). The key
   biomarker activity in this signature is increased cell surface levels of
   serum amyloid A (SAA) protein. Analysis of a large reference database (BioMAP
   Phenotypic Profile Reference Database) of >3400 drugs and chemicals tested in
   this assay identified 147 compounds exhibiting the signature at one or more
   concentrations. For some of these compounds, specific mechanisms could be
   implicated and include MEK inhibition, HDAC inhibition, glucocorticoid
   (GR)/mineralocorticoid (MR) receptor agonism, IL-6 pathway agonism, as well
   as modulation of mitochondrial NAD+/NADH ratios.
   
   To further characterize the mechanisms underlying this toxicity-associated
   signature, we took advantage of a second large reference database (BioPrint®
   Pharmacology Profile Database) comprised of in vitropharmacological profiles
   of drugs and chemicals screened against a broad range of targets (~148
   receptors, ion channels, enzymes and transporters). We evaluated the in
   vitropharmacology profiles for compounds exhibiting the phenotypic signature
   associated with cardiovascular toxicity(data was available for 85 of 147
   compounds). Target activities (in binding assays) enriched among the
   phenotypic actives include glucocorticoid receptor (GR), androgen receptor
   (AR), chloride channel (Cl-channel), ML2 (MT3), (5-Hydroxytryptamine receptor
   2B (5-HT2B), peripheral benzodiazepine receptor (BZD), MT1 and ML1. The
   identification of ML2 (MT3), also known as NAD(P)H quinone dehydrogenase 2 or
   NQO2, and MT1 receptors is interesting as these are receptors for melatonin.
   Melatonin has been reported to reduce blood pressure and also to reduce NAD+
   levels through effects on NAMPT (nicotinamide phosphoribosyltransferase).
   Recent studies have suggested that NAMPT may play a role in the pathogenesis
   of atherosclerosis in experimental mouse models. In humans, serum
   concentrations of NAMPT are an independent predictor of symptomatic carotid
   stenosis in patients undergoing carotid endarterectomy.
   
   These results show how the combined analysis of phenotypic and pharmacology
   profiling data can confirm and extend our understanding of potential
   mechanisms associated with the risk of cardiovascular toxicity. The pairing
   of target-based and phenotypic assays is an efficient and effective means to
   improve confidence in non-animal based screening of new drug leads for
   potential liabilities.

Back to Top


ASSAY DEVELOPMENT AND SCREENING

Track Chair(s): Joe McGivern, Ph.D., Amgen (USA) and Melissa Crisp, Ph.D., Eli
Lilly (USA)

ADVANCED IMAGING & HIGH-CONTENT ASSAYS

Session Chair: James Evans, Ph.D., Phenovista Biosciences (USA)

 * Applications of high content imaging in drug discovery using physiologically
   relevant models of disease
   Monica Chu, Phenovista
   
   Cell-based imaging assays are used extensively for drug discovery; however,
   challenges remain in the translation of in vitro data to in vivo outcome. We
   are focused on generating relevant and translatable high content imaging data
   from physiologically relevant cell models, including 3D structures. Case
   studies will be presented on the application of high content imaging assays
   to several complex, physiologically relevant cell-based models, including
   ex-vivo patient-derived xenografts for assessment of tumor/immune cell
   interactions in the tumor microenvironment; iPSC derived mini-brains for
   assessment of neuronal health, and cell painting in 3D organoids.

 * Deep, Single-Cell Analysis by Microscopy: Beyond Human Vision
   Anne Carpenter, Broad Institute of Harvard and MIT
   
   Microscopy images contain tremendous information about the state of cells,
   tissues, and organisms. We aim to go beyond measuring individual phenotypes
   that biologists already know are relevant to a particular disease. Instead,
   in a strategy called image-based profiling, we stain many cellular components
   and extract thousands of morphological features from each cell’s image, often
   using an assay called Cell Painting. We then harvest similarities in these
   “profiles” to identify, at a single-cell level, how diseases, drugs, and
   genes affect cells, which can uncover small molecules’ mechanism of action,
   discovering disease-associated phenotypes, identify the functional impact of
   disease-associated alleles and identify novel therapeutics.

 * Cell-Based Screening to Identify a Lead Humanised Antibody Drug Conjugate
   Siobhan Leonard, LifeArc
   
   Antibody-drug conjugates (ADCs) are being designed and used as highly
   targeted cancer therapies, with five approved by the FDA and over sixty in
   clinical trials. Building on the success of antibody therapies, ADCs enable
   highly specific delivery of a toxic payload to a target tumor cell. LifeArc
   has previous experience in the successful humanization of antibodies for
   clinical use, Keytruda, Entyvio, Actemra and Tysabri. In order to build upon
   this expertise and extend the therapeutic approaches, LifeArc has several ADC
   programmes in the oncology and non-oncology space. As part of this portfolio,
   the in-house capability has been established and several cell-based assays
   have been developed to identify candidate ADCs. A case study will be
   presented that outlines the screening process used to characterize the
   capacity of candidate antibodies to bind, internalize and induce cell death
   in HEK293 cells overexpressing the receptor of interest.
   
   Critical to the success of an ADC program is the development of effective
   methodologies to screen for candidate internalization to the lysosome, where
   the linker will be cleaved to release the attached cytotoxin. To facilitate
   high-throughput analysis of hybridoma supernatant and humanized variant
   internalization, LifeArc has developed robust cell-based assays with the
   IncuCyte S3 Live-Cell Analysis System and pH-sensitive dyes which fluoresce
   in acidic lysosomes and endosomes. Following rapid evaluation with the
   IncuCyte, deeper insight into the intracellular trafficking of promising
   candidates was gained with the IN Cell Analyzer 6500 HS high content analysis
   system. When coupled with intuitive analysis workflows to evaluate antibody
   co-localisation with the lysosome, the sophisticated sensitivity of this
   high-content imager has allowed further insight into the profile of promising
   drug candidates and has facilitated the validation of this receptor as an ADC
   target.

 * Cell Painting in Hit Discovery
   Charles-Hugues Lardeau, AstraZeneca
   
   Having historically looked at characterizing our compound collection for
   cytotoxicity and cytostaticity or performed the routine annotation of the
   AstraZeneca compound collection with frequent hitters, we are looking into
   alternative compound annotation solutions (e.g. imaging or transcriptomics
   output) and the added benefits they may provide. The Cell Painting imaging
   assay multiplexes six fluorescent stains to enable the visualization of up to
   eight cellular compartments in U2OS cells. We will present how we have
   established the Cell Painting imaging assay in 384-well plates and made it
   compatible with our CoLab automation platform. The use of a centrifugal plate
   washer afforded the reduction of staining solution needed and combined with a
   microvalve-based microplate dispenser allowed the miniaturization of the
   assay to 1536-well plates. Driven by sustainability, we have also explored
   the use of a microplate cleaning system to wash and reuse plates for this
   assay. Specific collections we have applied the Cell Painting imaging assay
   include a set of compounds also tested with a metabolomics output, compounds
   that are part of a phenotypic set or compounds from a nuisance compound set.
   On the analysis side, we will share our experiences using a traditional
   analysis pipeline (segmentation, working with ca. 1500 features) and the work
   we are planning to do using deep learning models to best exploit images and
   data generated. The latter part will touch on the work we have been doing to
   segment compartments from images.

BIOCHEMICAL AND BIOPHYSICAL ASSAYS

Session Chair: Amy Quinn, Ph.D., GlaxoSmithKline (USA)

 * Enzymology Framework and Assay Platform for Targeted Protein Degradation
   Optimization
   Stewart Fisher, C4 Therapeutics, Inc.
   
   Targeted protein degradation, through the use of heterobifunctional degraders
   that act as catalytic activators for an E3 ligase and target protein, has the
   potential to transform drug discovery. This talk will discuss the key
   structure: activity relationships that underpin degrader optimization,
   including the interpretation and interplay of both thermodynamic and kinetic
   elements that drive the catalytic turnover of targeted proteins. These data
   will be placed in context with an enzymology framework to characterize
   cellular degradation data the extension of these insights to pharmacodynamic
   modeling and predictions.

 * Mass Spectrometric Assay of METTL3/METTL14 Methyltransferase Activity
   Shane Buker, Accent Therapeutics
   
   A variety of covalent modifications of RNA have been identified and
   demonstrated to affect RNA processing, stability and translation. Methylation
   of adenosine at the N6 position (m6A) in mRNA is currently the most
   well-studied RNA modification and is catalyzed by the RNA methyltransferase
   complex METTL3/METTL14. Once generated, m6A can modulate mRNA splicing,
   export, localization, degradation and translation. Although potent and
   selective inhibitors exist for several members of the Type I
   S-adenosylmethionine (SAM)-dependent methyltransferase family, no inhibitors
   have been reported for METTL3/METTL14 to date. To facilitate drug discovery
   efforts, a sensitive and robust mass spectrometry-based assay for
   METTL3/METTL14 using self-assembled monolayer desorption/ionization (SAMDI)
   technology has been developed. The assay uses an 11-nucleotide
   single-stranded RNA compared to a previously reported 27-nucleotide
   substrate. IC50 values of mechanism-based inhibitors S-adenosylhomocysteine
   (SAH) and sinefungin (SFG) are comparable between the SAMDI and radiometric
   assays that use the same substrate. This work demonstrates the SAMDI
   technology is amenable to RNA substrates and can be used for high-throughput
   screening and compound characterization for RNA modifying enzymes.

 * Fragment-Based Target Screening - An Empirical Approach to Prioritising
   Targets; A Case Study on Antibacterials
   Peter Coombs, LifeArc
   
   In the post-genomic era, abundant functional genomic data is being generated
   and lists of potential new druggable targets are being analyzed across
   industry and academia. Bioinformatics and literature diligence can take us so
   far, but there are two key aspects to target selection that are best examined
   empirically: tractability and ligandability. We have developed a
   high-throughput fragment screening approach we call Fragment-Based Target
   Screening (FBTS) to examine this.
   
   The FBTS approach has been exemplified at LifeArc in its application to
   antibacterials. New antibacterial drugs are urgently needed to tackle the
   emerging crisis of multidrug-resistant infections. 50 potential targets were
   selected by bioinformatic analysis from a TraDIS dataset of essential genes.
   Our high-throughput expression and purification platform were used to test
   the targets for expression. Biophysical techniques were used to QC 38
   proteins and match them with a control ligand. Proteins were screened against
   a library of 1280 fragments on a Biacore 8K, then hits confirmed in
   dose-response experiments. Automated data processing and machine learning
   were applied to assign a ligandability score to each target & prioritize hit
   fragments for progression. This was combined with additional essentiality
   data & a progression assessment to compile a priority list of targets to
   advance into drug discovery projects.
   
   FBTS is accessible; fragment libraries and the equipment needed for protein
   production and fragment screening are not only in the domain of big pharma,
   and the cost and effort associated less daunting than that typically
   associated with other screening options such as HTS or DNA-encoded libraries.
   In addition, the approach lends itself to take advantage of the wealth of
   structural data from public databases and structural genomics consortia.
   Combined with novel functional genomic data, our empirical tractability and
   ligandability assessment allows high-quality targets to be prioritized for
   prosecution and improved chances of success.

 * Identification and Biophysical Characterization of STING Modulators
   Charles Lesburg, MSD
   
   The second messenger cyclic dinucleotide (CDN) cGAMP is produced by the cGAS
   protein in response to activation by cytoplasmic dsDNA. Upon recognition of
   cGAMP by the stimulator of interferon genes (STING) protein, STING undergoes
   a substantial conformational change which leads to downstream upregulation of
   proinflammatory cytokines. Modulation of the cGAS/STING pathway is therefore
   considered a promising route for the treatment of inflammatory diseases as
   well as a potential partner for immune-oncology therapies. This presentation
   will describe the identification and optimization of compounds that were
   found to antagonize STING signaling as well as the identification and
   conformational characterization of non-CDN STING agonists. Techniques
   employed include surface plasmon resonance and X-ray crystallography.

ADVANCED IN VITRO CELL CULTURE SYSTEMS

Session Chair: Virneliz Fernandez-Vega, B.S., Scripps Research (USA)

 * Pancreatic Cancer Patient-Derived Organoids as a Tool for Personalized
   Medicine
   Herve Tiriac, University of California, San Diego
   
   Pancreatic cancer is the most lethal common solid malignancy. Systemic
   therapies are often ineffective and predictive biomarkers to guide treatment
   are urgently needed. We generated a pancreatic cancer patient-derived
   organoid (PDO) library that recapitulates the mutational spectrum and
   transcriptional subtypes of primary pancreatic cancer. New driver oncogenes
   were nominated and transcriptomic analyses revealed unique clusters. PDOs
   exhibited heterogeneous responses to standard-of-care chemotherapeutics and
   investigational agents. In a case study manner, we find that PDO therapeutic
   profiles paralleled patient outcomes and that PDOs enable longitudinal
   assessment of chemo-sensitivity and evaluation of synchronous metastases. We
   derived organoid-based gene expression signatures of chemo-sensitivity that
   predicted improved responses for many patients to chemotherapy in both the
   adjuvant and advanced disease settings. Finally, we nominated alternative
   treatment strategies for chemo-refractory PDOs using targeted agent
   therapeutic profiling. We propose that combined molecular and therapeutic
   profiling of PDOs may predict clinical response and enable prospective
   therapeutic selection.

 * Drug Combination and Gene Network Analysis in 3D models of Brain and Pancreas
   Cancer towards Precision Medicine
   Tim Spicer, Scripps Research
   
   Molecular pathology approaches for clinical oncological care is routinely
   performed on cancer patients with recurrent or metastatic disease. While
   these “omic” diagnostics seemingly improved prognostication and prediction,
   some molecular 'signatures' are not useful in clinical practice because of
   their inability to independently validate treatment options. By nature,
   associations between genomic profiles and clinical response are correlative
   rather than mechanistic resulting in poor prediction for needed care.
   Advances in our lab, in combination with our academic and industry partners,
   has made possible in-vitro/ex-vivo 3 dimensional (3D) models of cancer
   biology for use in a rapid, highly miniaturized, and cost-effective fashion
   that permits direct drug response profiling to be generated in a phenotypic
   manner that is patient specific. By integrating genomic diagnostics with drug
   response testing a significant breakthrough toward advancing precision
   medicine, using tumor biopsies, is now technologically possible and is
   referred to as Precision Medicine Therapeutic Profiling. Glioblastomas and
   cancer of the pancreas represent two of the most lethal malignancies with
   survival typically less than two years from diagnosis. These models of
   malignancy combined with genetic profiling have been tested in 3D cultures to
   validate the best drug, or combination of drugs, for individualized care in a
   time frame that is meaningful to clinical application. It is hypothesized
   that the comprehensive data generated will afford physicians with a powerful
   new insight that is actionable for patient care.

 * New Innovation to Solve Unmet Needs: Implementing Human Induced Pluripotent
   Stem Cell-Derived Neural Spheroids as a Robust Screening Platform for
   Phenotypic-Based Central Nervous System Drug Discovery
   Oivin Guicherit, StemoniX
   
   The central nervous system (CNS)-based drug discovery has been hampered by a
   lack of relevant, high-throughput experimental platforms. Complex,
   three-dimensional (3D), experimental preparations with multiple cell types
   better represent the native, in vivo biology, thus providing relevant
   material for CNS investigations. Unfortunately, these preparations
   traditionally have not been able to support the throughput necessary for
   early-stage discovery programs. The ideal preparation would provide
   consistent native tissue function in high throughput plates. To meet this
   need, we have developed 96- and 384-well assay-ready, 3D neural spheroid
   platforms; each spheroid is composed of cortical glutamatergic and GABA-ergic
   neurons co-cultured with astrocytes to provide a more complex, biologically
   relevant, and predictive preparation in a high throughput platform for
   compound screening, safety evaluation, and toxicity studies.
   
   Whole-genome RNAseq profiling demonstrated neural tissue expression patterns,
   and high content imaging validated neuronal and astrocytic cell populations
   while showing highly reproducible spheroid size across both 96 and 384-well
   platforms. Functional neuronal activity was confirmed with MEA recordings and
   visualized under high-throughput conditions as robust spontaneous,
   synchronized calcium oscillations with consistent and reproducible baseline
   activity patterns across wells and plates. Functional circuitry was confirmed
   by challenging the system with specific ion channel and neurotransmitter
   receptor agonists and antagonists.
   
   To validate the capabilities of the platform for compound profiling and
   discovery, a library of 1622 FDA approved compounds were screened in single
   point at 10 µM final concentration examining Ca2+ oscillations as a
   functional phenotypic readout. The library included drugs covering a wide
   spectrum of targets such as CNS biology, oncology, cardiology,
   anti-inflammatory, immunology, neuropsychiatry and analgesia with DMSO as a
   vehicle control. Hits were identified as responses that were at least 3
   standard deviations from DMSO control responses. As expected, the highest
   number of hits were from targets associated with neuronal signaling
   (serotonin, dopamine, GABA, and adrenergic receptors), neural biology, and
   second messengers such as cAMP. Of note was the identification of several
   compounds that led to increases in peak count similar to that of 4-AP, a
   known pro-convulsant. The results validated a robust screening platform with
   a vehicle control standard deviation of ~9% across all plates and a Z’ score
   of 0.73 across the entire screen.
   
   In conclusion, performing a high-throughput functional screening assay on our
   human iPSC-derived 3D neural spheroid platform demonstrated the ability to
   identify a wide range of hits spanning multiple target areas. This model may
   serve as a phenotypic and target-based platform for overcoming traditional
   hurdles of CNS-based drug discovery and improving outcomes for novel
   CNS-targeted drug discovery and development efforts. Moreover, the model can
   be created from both wild type and diseased individuals, providing relevant
   human platforms for disease-specific drug discovery.

 * Maximizing the Value of Cancer Drug Screening in Multicellular Tumor Spheroid
   Cultures – Are You Analyzing Your 3D Tumor Models Appropriately?
   Paul Johnston, University of Pittsburgh Dept. Pharmaceutical Sci.
   
   Historically, cancer drug leads are identified in high-throughput screening
   (HTS) growth inhibition assays performed in tumor cell line panels maintained
   and assayed in 2-dimensional cultures. However, the overall probability for
   success in oncology clinical trials is a dismal 3.4%. To improve clinical
   development success rates for solid tumors, more physiologically relevant in
   vitro 3-dimensional models are being deployed in lead generation to identify
   better cancer drug candidates. Multicellular tumor spheroids (MCTSs) resemble
   avascular tumor nodules, micro-metastases or the intervascular regions of
   large solid tumors concerning morphology, volume growth kinetics, and form
   diverse microenvironments due to gradients of nutrient distribution and
   oxygen concentration. Head and neck cancers (HNC) are the 8th leading cause
   of cancer worldwide and in 2019 it’s projected that 53,000 people in the USA
   will develop oral cavity or pharynx cancer and 10,860 will die of these
   cancers. Seven drugs are approved for HNC therapy, but only 10-25% of
   patients respond to single-agent therapy, and 5-year survival and/or cure
   rates have not improved. Although pembrolizumab (Keytruda®) was well
   tolerated in patients with recurrent or metastatic HNC and produced
   clinically relevant antitumor activity, only 16% of patients responded to
   treatment. The low response rates and limited efficacy of HNC drugs
   underscore the need to discover new and effective therapies. We have
   developed methods to characterize HNC MCTS morphologies, viability and growth
   phenotypes and to conduct cancer drug HTS. In a total of 95 pairwise cancer
   drug x HNC cell line experiments, only 35.8% of MCTS cultures exhibited a
   concentration-dependent growth inhibitory response using metabolic viability
   reagents, and only 24.4% produced ≥50% reduction in Calcein AM live cell
   staining. In contrast, 67.8% increased ethidium homodimer dead cell staining
   by ≥50% and 89.5% altered ≥1 morphological feature; size, shape/perimeter or
   density/compactness. These data demonstrate that multiple analysis methods
   are required to accurately assess the impact of cancer drugs on HNC MCTS
   cultures and to maximize the value of these physiologically relevant tumor
   cultures.

DEVELOPMENT OF CELLULAR ASSAYS TO UNDERSTAND PROTEIN HOMEOSTASIS

Session Chair: Dane Mohl, Ph.D., Amgen (USA)

 * Novel Strategies for Oncoprotein Degradation
   Willem Den Besten, Amgen
   
   Targeted protein degradation has the potential to open the door to
   therapeutic targets previously deemed undruggable. In this talk, I will
   present the characterization of two ligase ligands and show how target
   degradation coupled with modulation of ligase biology leads to increased
   cellular efficacy. I will also share results on a new method for inducing the
   degradation of an ubiquitin ligase.

 * Quantitative Live Cellular Assays for Screening Degradation Compounds and
   their Mechanism of Action
   Kristin Riching, Promega Corporation
   
   A new generation of heterobifunctional small molecules, termed PROTACs, holds
   significant therapeutic potential by inducing degradation of target proteins.
   These compounds consist of two binding regions separated via a linker: one
   that specifically binds to the target protein, and the other that directly
   recruits E3 ligase machinery, resulting in ubiquitination and degradation of
   the target. Characterizing PROTAC degradation efficacy represents a
   significant challenge, both in terms of understanding the individual
   mechanistic processes that control whether degradation will result, as well
   as the ability to screen for target protein loss in high throughput fashion.
   Here, we present a live-cell, luminescence-based technology platform that
   enables characterization and screening of PROTAC compounds and their
   mechanism of action using either ectopic or endogenous target expression
   formats. We employ CRISPR/Cas9 endogenous tagging of target proteins with the
   small peptide, HiBiT, which has a high affinity for and can complement the
   LgBiT protein to produce NanoBiT luminescence. This allows for sensitive
   detection of endogenous protein levels in living cells, and can also serve as
   a BRET energy donor to study protein: protein or protein: small molecule
   interactions. Using this combinatorial approach, we demonstrate the ability
   to measure permeability effects and binding affinities of PROTAC compounds to
   both target and E3 ligase, as well as monitor the kinetics of the subsequent
   ternary complex (target:PROTAC: E3 ligase) formation, target ubiquitination
   and recruitment to the proteasome in live cells. We further show the power of
   this technology in extended kinetic monitoring of endogenous target protein
   levels, quantification of key degradation parameters for rank-ordering,
   correlation of these parameters to the precise MOA and the application of
   these approaches for HTS. This comprehensive technology platform enables
   rapid, simple and robust screening of functional degrader compounds,
   ultimately aiding chemical design strategies for the optimization of new
   therapeutic PROTACs.

 * Cellular Assays Targeting Two Mutation Classes Causing Cystic Fibrosis:
   Through (1) Protein Misfolding or (2) Premature Translational Termination
   Feng Liang, Cystic Fibrosis Foundation
   
   Cystic fibrosis (CF) is a disease caused by mutations in the gene coding for
   the cystic fibrosis transmembrane conductance regulator (CFTR), a chloride
   channel. Mutations are classified into six classes with phenotypes from no
   CFTR protein synthesis to misfolding and/or functional defects.
   
   Over the past seven years, the FDA approved several novel small molecules
   that partially correct defects of different mutation classes of CFTR. This
   has triggered broad efforts to find better and/or different small molecule
   modulators that address even more CF disease-causing mutations. Here we
   present screening assays for two classes of CFTR variants: (1) F508del
   (causing protein misfolding and severely impaired cellular trafficking) and
   (2) premature termination codon (PTC) mutations, resulting in stop codons in
   the open reading frame of CFTR and no functional expression. Assays need to
   address the primary defects of these specific mutation types. A differential
   screening approach allows the discovery of class-specific hit molecules.
   
   CFTR F508del leads to (1) misfolding of the nucleotide-binding domain 1
   (NBD1) of CFTR and (2) perturbs normal interdomain interaction in the CFTR
   protein. An efficient therapy needs to address both protein folding defects
   for CFTR for the rescue of CFTR functional expression. Suppressing one defect
   may allow identification of modulators of the 2nd defect. Thus, using
   specific suppressor mutations (R555K to restore NBD1 folding or R1070W to
   rescue domain-domain interactions, allelic screens were developed to enrich
   for small molecules that preferentially modulate interdomain interactions or
   NBD1 folding, respectively. The phenotypic screen relies on mammalian cells
   expressing CFTR F508del with the suppressor mutations and a reporter gene
   fused into an extracellular loop of CFTR. Hits from the two assays were
   further tested for complementary effects on the trafficking rescue of CFTR
   F508del.
   
   A different class of CFTR mutations are PTC variants (about 170 reported)
   that cannot be treated with available medicines. During CFTR protein
   synthesis, the interaction of the ribosome with the PTC (UAA, UAG, or UGA)
   terminates protein translation. Furthermore, when the ribosome stalls at a
   PTC, translation-coupled RNA surveillance triggers the nonsense-mediated mRNA
   decay (NMD) pathway, resulting in a reduction of CFTR mRNA levels. Therefore,
   an effective therapy for CFTR PTC variants needs to address both premature
   translation termination and reduced CFTR mRNA. Cell-based assays to assess
   translational readthrough of PTCs have been developed based on either a
   reporter or the native CFTR gene. RT-qPCR of CFTR mRNA is utilized to monitor
   anti-NMD effects. Our data support the concept that combining readthrough
   modulators and NMD inhibitors may lead to more effective therapy.
   
   The CF phenotypes for the above two classes of CFTR mutations derive from
   defects in different stages of CFTR biogenesis. Specific types of mutations
   require different screens for the identification of mutation class-specific
   disease modulators.

 * Cellular Thermal Shift Assays in High-Throughput: A 1536-Well Cellular Target
   Engagement Assay for Drug Discovery
   Lorena Kallal, GlaxoSmithKline
   
   Thermal shift assays (TSA) reveal changes in protein structure upon binding
   to small molecules due to a resultant change in the thermal melting
   temperature of the protein. Experimentally, this change in melt temperature
   can be measured by exposure of the protein to a temperature gradient,
   followed by quantification of the protein level or activity at each
   temperature. Originally, protein thermal shift experiments were performed
   with purified protein samples, but recently the TSA was reported in a
   cellular context and the cellular thermal shift assay (CETSA) was born. We
   have combined CETSA with a high-throughput protein detection method to
   increase the throughput of the assay since traditional protein detection
   methods such as western blots are low throughput. To develop high-throughput
   1536-well CETSA, we used a protein reporter system in a homogeneous
   (additions only, no wash) assay format. We have successfully utilized this
   assay to characterize compounds in dose-response curves for drug discovery
   programs at GSK. This method can also be applied to identify hits in high
   throughput screening. Assay parameters optimized included target expression
   level, the number of detection reagents added after thermal melting, plate
   type and thermal melt methodologies. Uses and applications in drug discovery
   will be presented.

APPLICATIONS OF CRISPR TECHNOLOGY IN DRUG DISCOVERY

Session Chair: Melissa Crisp, Ph.D., Eli Lilly (USA)

 * Quantitating Endogenous Protein Dynamics with a Bioluminescent Peptide Tag
   Marie Schwinn, Promega Corporation
   
   There are an estimated 3,000 human genes that constitute the “druggable
   genome.” However, only a small percentage of proteins coded by these genes
   are the focus of drug discovery programs. One barrier in investigating these
   understudied targets is the lack of easily implemented and scalable methods
   for assaying proteins. The two principal techniques for analyzing proteins
   are immuno-detection and mass spectrometry. They offer the advantage of
   generating data from endogenously expressed proteins. However, these methods
   are limited by the lack of protein-specific reagents, sensitivity, and HTS
   compatibility. This prompted us to develop a workflow for studying endogenous
   proteins that were both easy to use and scalable. In recent years, CRISPR
   technology has been utilized to integrate reporters into host genomes, such
   that cellular proteins can be monitored in real-time through detection of the
   reporter fusion. CRISPR-mediated knockin of the HiBiT luminescent peptide
   reporter has been demonstrated on a small-scale using a cloning-free
   workflow. The high sensitivity and dynamic range associated with HiBiT make
   it suitable to study most cellular proteins across a range of expression
   levels. Thus, we wanted to determine if CRISPR-mediated HiBiT tagging would
   provide an approach to rapidly tag any protein in the human proteome. To
   explore this strategy, a diverse set of proteins representing a broad range
   of functions and biophysical properties were targeted for tagging with the
   HiBiT luminescent peptide tag. The majority of the selected targets showed
   successful integration and expression of functional fusion protein. Given the
   high success rate in this initial experiment, we investigated if this
   strategy could be used for developing an HTS-compatible assay for an entire
   protein family. For this purpose, the cyclin-dependent kinase (CDK) family
   was targeted for HiBiT tagging and then used to quantitate CDK-specific
   target engagement. Although the majority of edited CDK-HiBiT cell lines
   displayed compound pharmacology similar to what was observed in
   over-expression-based models, several differences were found which suggests
   that endogenous models may provide more accurate information on compound
   activity. In summary, CRISPR-mediated tagging of endogenous proteins with
   HiBiT represents an easy and scalable strategy for studying endogenous
   proteins which enables the analysis of proteins in their appropriate
   physiological context.

 * A 384-Well Workflow to Execute an Arrayed CRISPR-Cas9 Gene Editing Screen in
   T-Cells
   Sapna Desai, GlaxoSmithKline
   
   Functional genomics approaches to identify novel therapeutic targets are
   rapidly gaining traction. Arrayed screening for the phenotypes resulting from
   gene-knockouts using CRISPR-Cas9 technology can yield results rapidly, with
   very little need for target deconvolution. Data can be further enhanced by
   the selection of disease-relevant primary cells.
   
   We have developed a high-efficiency, arrayed genome-editing screen in primary
   CD4+ T cells using CRISPR–Cas9 for the identification of genes associated
   with cytokine release. T-cells are isolated, purified and expanded before
   genome editing occurs via nucleofection. A 384-well nucleofector is used to
   deliver RNP complexes consisting of guide RNA (gRNA), transactivating CRISPR
   RNA (tracrRNA) and Cas9 enzyme. Edited cells are rested and activated before
   being utilized in downstream assays capturing multi-cytokine release and cell
   viability.
   
   The development of miniaturized, robust nucleofection protocols and assays
   for T-cell screening allows integration of this challenging cell-type onto
   well-established liquid handling platforms and demonstrates the potential of
   genome-wide arrayed CRISPR-Cas9 screening of primary cells in a screening
   environment.

 * Development and Implementation of High-Throughput Cellular Protein Stability
   Assays for Evaluation of Target Engagement at Early Stages of Screening
   Projects
   John Holleran, Sanford Burnham Prebys Medical Discovery Institute
   
   Cellular protein thermal stability provides a powerful method for assessing
   compound target engagement. Recently, there have been several high-throughput
   384 well assay compatible detection formats published for cellular protein
   thermal stability using commercial luminescence complementation systems as
   well as homogenous antibody sandwich AlphaLISA assays. Similarly, we have
   utilized these detection formats and successfully developed and employed
   high-throughput format assays for a large number of diverse drug discovery
   projects ongoing at the SBP Prebys Center. In addition to serving the primary
   goal of assessing the target engagement, these efforts have provided in-depth
   knowledge of diverse detection systems, such as Promega HiBiT, DiscoveRx ePL,
   AlphaLISA and the classical approach relying on SDS-PAGE-Western Blot
   detection. In addition to monitoring the thermal stability of intracellular
   proteins, we successfully employed these same detection approaches to assess
   small-molecule effects on steady-state protein level or localization in the
   cell. Not only do these approaches enable reporting on direct target binding,
   but they also monitor the effects of small molecules on the protein target
   interactome governing homeostasis. This enables the identification of
   druggable partners from the entire target interaction network. Along with
   small molecule screening, all these methods provide unique and powerful tools
   to study targets of interest in their native state which exposes valuable
   information about the effects of cellular background and extracellular
   environment. From these studies, we have gleaned insight into the cellular
   and context-specific target regulation and potential biological relevance of
   identified hits. We have successfully utilized both exogenous expression and
   CRISPR knock-in of detection tracers or AlphaLISA detection to assess protein
   stability of endogenous proteins learning advantages and disadvantages of
   each approach. Side-by-side assessment of these approaches helped to develop
   a decision tree for selecting the most appropriate approach for each new
   project and target.

 * The Development and Application of a Whole Genome Arrayed CRISPR Screening
   Platform for Target Discovery and Mechanism of Action Investigation
   Douglas Ross-Thriepland, AstraZeneca
   
   The identification of novel therapeutic targets that translate into clinical
   successes is needed now more than ever to deliver life-changing medicines to
   patients across disease areas; from drug resistance in oncology to
   cardiovascular and respiratory disease. In this effort, the unbiased
   identification of targets through perturbation at the gene level is not new.
   However, the CRISPR/Cas9 revolution has enabled us to achieve this with
   higher efficiency, reduced off-target effects and has enabled new modes of
   perturbation such as gene activation (CRISPRa) and SNP mutation (base
   editing). We have used this technology to build a Target Discovery platform
   encompassing both pooled and arrayed screening techniques that, taken
   together, allow us to probe a wide range of biology, from the slow onset of
   drug resistance seen in oncology (pooled), through to understanding what role
   genes play in signaling and cell response (arrayed). Here we present the
   development and application of our Arrayed CRISPR Screening component of this
   platform. Using CRISPR libraries comprised of synthetic gRNAs arrayed into a
   “one-gene-per-well” format we demonstrate the high-efficiency of both genes
   knock out (CRISPRn) and gene activation (CRISPRa) at whole genome-scale in
   cell-based assays. Coupled with the generation of high-quality Cas9
   expressing cells through the ObLiGaRe insertion of an inducible Cas9
   expression cassettes (ODin), we show that CRISPR/Cas9 is a powerful and
   robust technology for arrayed screening. By combining this platform with
   high-content imaging and multivariate analysis technology we have been able
   to interrogate the phenotype resulting from gene perturbation to a much
   greater depth. This has significantly improved how we are able to rank,
   triage and progress hits into target validation and into the clinic. Here we
   demonstrate the application of this platform for the unbiased discovery of
   new therapeutic targets with two case studies that exemplify the capability
   and how it has impacted our new target pipeline in both oncology and advanced
   drug delivery.

OPTIMIZATION OF ASSAYS FOR CHALLENGING TARGETS FOR LEAD DISCOVERY AND SCREENING

Session Chair: Julie Conkright-Fincham, Ph.D., Stowers Institute for Medical
Research (USA)

 * Functional Proteome Array Screening Strategies for Biomarker Discovery
   Joshua LaBaer, The Biodesign Institute, ASU
   
   Self-assembling protein microarrays can be used to study protein-protein
   interactions, protein-drug interactions, search for enzyme substrates and as
   tools to search for disease biomarkers. In particular, recent experiments
   have focused on using these protein microarrays to search for antibody
   responses in patients with cancer, autoimmune and infectious diseases. This
   approach has led to the first CLIA-certified blood test for the early
   detection of breast cancer, Videssa™. Recent work has focused on using the
   arrays to explore the post-translational modification of proteins and their
   role in producing neoantigens in disease.

 * NanoClick Assay: A High-Throughput, Target-Agnostic Cell Permeability Assay
   that Combines NanoBRET Technology with Intracellular Click Chemistry
   Andrea Peier, MSD
   
   Macrocyclic peptides open new opportunities to target intracellular
   protein-protein interactions (PPIs) that are often considered non-druggable
   by traditional small molecules. Specifically, peptides have the potential to
   bind to highly expansive binding surfaces (orthosteric blocking) of such PPIs
   and/or other unique allosteric binding sites. However, their clinical
   development may be limited by their ability to efficiently penetrate cells to
   modulate their cognate PPI targets. The ability to have a predictive,
   high-throughput assay to assess cell permeability is a critical tool to
   support peptide drug discovery programs.
   
   We developed a high throughput, quantitative, target-agnostic cell
   permeability assay that essentially measures the cumulative cytosolic
   exposure of a peptide in a concentration-dependent manner. The assay has been
   named NanoClick as it combines in-cell Click chemistry and monitoring of a
   NanoBRET signal in cells. The assay is based on cellular expression of the
   NanoLuc-HaloTag system and relies on the Click reaction of azide-containing
   peptides with DiBac-chloroalkane (CA) anchored to the HaloTag. The subsequent
   introduction of an azido-dye followed by the NanoLuc substrate allows the
   detection of a BRET signal that is reduced by the presence of Click-reactive
   peptides in the cytosol. The readout can be expressed as a permeability ratio
   of EC50s when compared to the response of a low permeability control.
   
   We validated the assay using known cell-penetrating peptides and were further
   able to demonstrate correlations to cellular activity using a p53/MDM2 model
   system. The assay has been applied across multiple programs and has been used
   to guide and establish structure-permeability relationships in the
   optimization of macrocyclic peptides for cellular potency across
   intracellular PPI target programs.

 * Building Toolkits for the Orphan Kinome
   Laurie Parker, University of Minnesota
   
   Protein phosphorylation by kinases is a major mechanism of cell signaling and
   is involved in almost all aspects of cell biology. Kinase dysregulation is a
   key factor in diseases like cancer, and kinases are one of the major drug
   targets in oncology. However, despite decades of research and billions of
   dollars in drug discovery efforts on kinases, relatively few are well
   characterized. The majority of the ~90 tyrosine kinases are considered
   “orphans,” for which few to no substrates, and thus few details about
   biological pathways and roles, are known. Without substrates to use as
   activity probes, inhibitors for use as tool compounds or potential
   therapeutics cannot be discovered. We have developed a strategy to
   incorporate empirically-determined substrate profiling data into our
   KINATEST-ID bioinformatics pipeline to efficiently tackle the orphan kinase
   problem, determine substrate preferences and design novel substrate tools.
   Protease-digested peptides from cell lysates are stripped of pre-existing
   phosphates, then re-phosphorylated with a kinase of interest. The resulting
   phosphopeptides are enriched and analyzed using mass spectrometry.
   Phosphopeptide sequences are extracted from the peptide ID list and funneled
   through the KINATEST-ID pipeline using a set of scripts implemented in the
   open-source user interface GalaxyP, to define substrate sequence preferences
   and propose candidate optimal substrate peptides. Those are then synthesized
   and tested for phosphorylation efficiency by the target kinase. Using this
   approach, we have characterized substrate preferences for several
   understudied kinases for which few validated substrates were known, including
   FLT3 and two clinically relevant mutants, and BTK. Current and future efforts
   are to broaden the scope of kinases characterized using this streamlined
   phosphoproteomics/bioinformatics pipeline and proceed with systematically
   defining substrate information and developing novel tools for other orphaned
   kinases in the kinome.

 * Hybridization Chain Reaction for Single-Cell Visualization of RNA in
   High-Content Imaging Assays
   Gianluca Pegoraro, National Cancer Institute
   
   The precise regulation of gene expression programs is responsible for the
   establishment and maintenance of cell, tissue and organ identity, for
   cellular responses to signaling cues and injuries, and, when disrupted or
   rewired, for diseases such as cancer and inflammation. Measuring gene
   expression in high-throughput assays often requires reporter cell line
   engineering, or using antibodies against endogenous protein markers, which
   involves a lengthy development process, and can also suffer from
   batch-to-batch variation. On the other hand, single-molecule RNA Fluorescence
   In Situ Hybridization (smRNA-FISH) detects endogenous transcripts, and is
   based on DNA oligonucleotide probes that can be rapidly designed in silico,
   chemically synthesized, tested, and scaled up. For these reasons, smRNA-FISH
   has the potential to be a useful additional tool for High-Content Imaging
   (HCI) in chemical or functional genetics screens for the identification of
   gene expression regulatory pathways. However, visualization of RNA at the
   single-cell level via smRNA-FISH has not been optimized for HCI assays. To
   address these limitations, we adapted the single-step, enzyme-free RNA
   Hybridization Chain Reaction (RNA HCR) to a 384-well format using an HCI
   platform. First, we used RNA HCR probes against IFIT3, an
   interferon-stimulated gene (ISG), to demonstrate that high-throughput RNA HCR
   can quantitatively measure gene expression changes at the single-cell level
   in a 384-well format. As a proof of principle, we performed a focused RNAi
   screen against 521 human genes involved in epigenetics regulation to identify
   novel factors mediating the transcriptional response to interferon-γ. The
   results of this primary screen suggest that multiple components of the MOF
   acetylase complex are involved in the upregulation of IFIT3 upon interferon
   stimulation. Finally, we applied high-throughput RNA HCR in other HCI assays
   to measure expression levels of specific mRNA splicing isoforms of the FGFR2
   gene, to monitor the effect of steroid treatment on the expression of
   inflammation regulators in primary human monocytes, and to determine the
   effect of steroid treatment on a variety of GR-responsive genes in mouse
   cells. Altogether, these results indicate that RNA HCR can be miniaturized in
   384-well assays to semi-quantitatively detect several endogenous RNA species
   via HCI in physiologically relevant systems, at the single-cell level, and in
   a medium- to high-throughput format. In the future, we expect that
   high-throughput RNA HCR will be useful for the discovery and validation of
   diverse targets regulating gene expression.

Back to Top


AUTOMATION AND HIGH-THROUGHPUT TECHNOLOGIES

TRACK CHAIR(S): SAM MICHAEL, NIH (USA) AND HELEN PLANT, B.SC., ASTRAZENECA (UK)

ADVANCED IMAGING TECHNOLOGIES TO BRIDGE THE GAP BETWEEN HIGH-CONTENT AND
HIGH-THROUGHPUT

Session Chair: James Pilling, M.Sc., AstraZeneca (UK)

 * Revolutionizing Cellular Screening with Artificial Intelligence-Driven
   Label-Free Imaging
   Adam Corrigan, AstraZeneca
   
   The primary reasons for drug failure in the clinic are a lack of efficacy and
   safety. Therefore, to drive a better understanding of disease biology and
   improve the translation, cellular imaging assays in early discovery need to
   be increasingly complex, utilizing multiple biomarkers to label several
   proteins in a pathway and to quantify multiple sub-populations.
   
   The field of image analysis has been transformed by the explosion of machine
   learning and AI methods, and we are now leveraging recent developments to
   maximize the information we get from imaging data and enable new experimental
   approaches. A key limitation of machine learning, and particularly deep
   learning models is the requirement for large amounts of annotated training
   data. We have developed an active learning framework for efficient training
   data generation, alongside unsupervised phenotype discovery approaches, to
   build models that can quantify the full complexity of cellular screening
   data.
   
   We are also integrating label-free phase-contrast imaging into our cellular
   screens. A large amount of information on cellular morphology is contained in
   the phase-contrast images, which do not take up a fluorescent color channel,
   but the human interpretation is very difficult. By training a deep neural
   network to find and segment nuclei and cells from phase-contrast images
   alone, nuclear and cell markers are no longer required. This allows multiple
   biomarkers to be combined into a single screen, enabling more complex biology
   for less cost.
   
   In addition to segmentation, we have shown that standard readouts such as
   cell division and cell death can be predicted from the label-free images,
   opening the possibility for digital multiplexing of a wide range of
   biomarkers, in living cells, and without expensive cell engineering.
   Combining this method with the high-content Cell Painting assay, we are
   learning how to extract meaningful biological features from label-free
   images, which can then be used to re-interrogate existing screening data for
   new insights. This approach is straightforwardly integrated with existing
   workflows and is revolutionizing the questions being asked through cellular
   screening.

 * Applications of Image-Based Artificial Intelligence in Drug Discovery and
   Safety Testing
   Mahnaz Maddah, Dana Solutions LLC
   
   Discovering effective drugs and demonstrating their safety are significant
   challenges facing the pharmaceutical industry, due to the high costs of
   development, long lead times, and low success rates of late-stage clinical
   trials. There is a need for new tools and technologies to help identify safe
   and effective drugs during the early stages of development.
   
   Over the last decade, there has been significant progress in using human
   induced pluripotent stem cells (hiPSCs) for modeling of human disease, drug
   screening, and toxicity testing. Numerous studies have demonstrated that
   these cells have physiologically relevant characteristics and can be used for
   preclinical testing of new drugs using high-throughput assays. In such
   assays, image and signal analysis algorithms are used to generate
   quantitative measurements that relate to cell degradation, death, or changes
   in function. Such approaches may be missing subtle changes that are not
   easily visualized, are too complex to measure with traditional data analysis
   methods, and/or suffer from lack of consistent quality control metrics on the
   input data.
   
   Artificial intelligence (AI) techniques and specifically deep convolutional
   neural networks are perfectly suited to address the challenges of these
   high-throughput assays by analyzing large amounts of imaging data robustly
   and with a level of sensitivity that has not been previously possible. We
   present case studies for using AI for high-throughput image-based phenotypic
   screening, toxicity testing, and quality control. First, we present data from
   a drug discovery program for dilated cardiomyopathy using high-throughput
   imaging of sarcomere structure in stem cell-derived cardiomyocytes. We were
   able to build disease models with high accuracy, which were then deployed to
   identify small molecules that showed to reverse the disease phenotype. The
   identified small molecules were further validated with functional assays and
   preclinical mouse studies. Second, we present data from a pilot toxicity
   testing study using stem cell-derived cardiomyocytes. Our novel image-based
   AI method was successful in capturing dose-dependent structural changes on a
   panel of drugs with known cardiotoxicity profiles, while no change was
   detected for the negative control. The detected structural changes correlated
   strongly with contractility. Finally, we present data from a pilot quality
   control study using current-trace signals from a patch-clamp instrument. We
   successfully built an AI model that can accurately classify signals as good
   versus poor quality, which enables automated and consistent filtering of data
   during high-throughput experiments.

 * Optical Pooled Screens in Human Cells
   Avtar Singh, The Broad Institute
   
   Pooled genetic screens have been critical for the systematic identification
   of genes underlying cellular processes, but have thus far been limited to
   phenotypes defined by cellular enrichment or comparatively low-throughput
   single-cell molecular profiling. We have developed a method to make pooled
   libraries compatible with the rich set of spatially and temporally resolved
   phenotypes accessible to high-content microscopy by using targeted in situ
   sequencing to demultiplex genetic perturbations. We applied this technology
   to screen 952 genes for involvement in NF-κB signaling by imaging p65 nuclear
   translocation and relaxation, recovering most canonical pathway members and
   identifying novel candidate regulators of IL-1Β/TNFα-stimulated immune
   responses. We are currently piloting applications with a range of optical
   assays and cell models and expect that pooled optical screens will have broad
   utility in identifying genetic components, analyzing genetic circuits, and
   interrogating disease variants.

 * High-Throughput Single-Cell Imaging and Advanced MachineLlearning Supported
   Image Analysis of Primary Tumors Enables Anti-Cancer Therapy Development
   Gregory Vladimer, Allcyte
   
   The ability to perform high-content screening in a high-throughput fashion is
   routinely limited to cell lines and other explant model systems, however,
   there is a risk that these may not be fully representative of the in vivo
   environment due to culture adaptation or the lack of multi-lineage cell
   types. The ability to gather high-content data directly from primary samples,
   however, both direct from blood and bone marrow, metastasized cancers and
   dissociated solid tumor, without cell outgrowth or selection in a method
   amenable to laboratory automation can be a more direct system. Further, by
   combining imaging of these primary sample with an adaptable analysis
   pipelines robust to micro-aggregates, especially formed in solid tumor biopsy
   homogenates, vastly different cell shapes and sizes, and that can ultimately
   harness the features from each cell can become a powerful means to study drug
   response in a variety of indications using model systems directly derived
   from the patient. This methodology has been used to prioritize therapy for
   late-stage patients with hematological cancers in a basket trial (Snijder &
   Vladimer et al 2017, Lancet Hematology), has been integrated with genetic
   data to further uncover biological understanding and clinical synergy options
   (Schmidl & Vladimer et al 2019, Nat Chem Bio). Here, this talk will
   specifically focus on the details of the computational framework, including
   supervised and unsupervised machine learning approaches for cell
   identification and feature extraction, and other aspects of necessary
   infrastructure including cloud-deployment that is used to, in very
   high-throughput, quantify single-cell phenotypes form primary material from
   cancer patients for drug discovery. Further, the use case of understanding
   single-cell phenotypes after drug screening, both in single-cell suspensions
   and in micro-aggregate multi-cell / 3D environments, will be highlighted.

SCREENING & PROFILING AT HIGHER THROUGHPUT USING PHYSIOLOGICALLY RELEVANT CELL
MODELS

Session Chair: Roger Clark, B.Sc., Charles River Laboratories (UK)

 * A Systematic Medium-Scale Comparative Study of 2D Vs. 3D Models Using
   High-Content Imaging Approaches
   Thierry Dorval, Institut De Recherches Servier
   
   The development of new pharmaceutical drugs is an expensive and high-risk
   endeavor for the pharmaceutical industry. Major advances in physiologically
   relevant in vitro cellular assays such as three-dimensional models, induced
   pluripotent stem cells, organ-on-chip are expected to provide a better
   ability to predict therapeutic response, hence, reducing clinical attrition.
   Unlike high-throughput screening, high-content screening combines automated
   fluorescence microscopy with quantitative image analysis allowing phenotypic
   multiparametric readouts such as cell viability, DNA damage or mitochondria
   structure among many others. This approach is particularly well suited for
   complex or partially characterized targets. Moreover, in the oncology field,
   it has been shown that compound efficacy could be dramatically modulated in
   3D models.
   
   In this context, we are aiming to perform a medium scale screening campaign
   using a chemically diverse compounds collection on a cell line derived from
   non-small cell lung cancer with a specific mutation both in 2D and 3D models.
   Using high-content imaging approaches, a large set of parameters will be
   extracted, leading to a better characterization of various toxicity
   mechanisms of actions.
   
   To provide robust comparable results between cellular models, a specific
   subset of compounds was selected and screened in dose responses during the
   assay development workflow. The analysis of this rich set of complex data
   provided an opportunity to improve the rest of the screening campaign.
   
   Hits obtained from both screens will be classified, compared and validated in
   dose responses for a better understanding of the difference induced by the
   use of a 3D model combined with high-content imaging. Ultimately this could
   help assess the relevance of the 3D model in drug discovery in oncology.

 * A Novel Multiplexed uHTS and uHCS (MuHTCS) Platform in a 1536-Well Format for
   Chemical Biology Screening Using 3D Patient-Derived Cancer Organoids
   Yuhong Du, Emory Chemical Biology Discovery Center
   
   The current effort to grow human tissues as 3D “organoids” for cancer
   research aims to recapitulate 3D architecture of tumors in an in vitro
   environment for cancer biology studies and therapeutic development. However,
   due to various technical challenges, primary 3D organoid culture has not been
   widely used in a high-throughput screening (HTS) format for chemical
   screening. Here, we report the miniaturization and development of a
   multiplexed uHTS and uHCS (MuHTCS) organoid culturing platform for effective
   compound screening in a 1536-well format. Using pancreatic patient
   tumor-derived organoids as a model system, we optimized the 3D organoid
   culturing conditions with extracellular matrix (ECM). The growth of organoids
   was monitored by automated imaging. We further developed a multiplexed
   screening platform to simultaneously monitor the effect of compounds on the
   growth of organoids for ultraHTS (uHTS) and on the morphological change of
   organoids for ultra-high-content screening (uHCS) in a 1536-well plate. The
   MuHTCS assay has achieved Z’ > 0.5 and signal-to-background (S/B) > 6. A
   pilot screening of ~2000 FDA approved and the bioactive compound libraries
   have validated the assay for screening. Our data have demonstrated that it is
   feasible to utilize miniaturized 3D cancer organoids for large scale compound
   screening. The optimized MuHTC platform provides an efficient approach to
   accelerate 3-D organoids-enabled screening for drug discovery.

 * Acute Myeloid Leukemia Drug Sensitivity Testing Using Patient-Derived Cells
   in 1536 Format
   Lynn Rasmussen, Southern Research
   
   In practice, the choice of which drug to prescribe for an individual patient
   is often made without any information on how that individual will respond to
   a specific drug. The field of precision medicine is attempting to provide
   data to fill that information gap to match the patient with the most
   effective treatment for that individual. To try to fill that gap for AML
   patients we have developed a drug screening process using a panel of FDA
   approved drugs with patient-derived leukemia cells. Because the
   patient-derived cells are an extremely limited resource, a 1536-well assay
   format was developed to maximize the amount of data that could be generated
   for each patient. The process of developing this assay will be discussed,
   including the technical challenges, their solutions and the equipment choices
   used to achieve a reliable HTS format screening protocol.

 * Automating a CRISPR based rescue screen for Alzheimer’s phenotypes in iPSC
   derived neurons
   Shushant Jain, Charles River
   
   Alzheimer’s disease (AD) is a complex disorder with increasing prevalence and
   socio-economic burden. However, the majority of strategies aimed at
   identifying therapies for AD have been focused on targeting Abeta or TAU,
   which make up the plaques and tangles respectively commonly found in people
   with AD. The continued failure of the drug discovery process and the
   accompanying trials against these targets have necessitated more and better
   options for therapeutic intervention. Using multi-parametric high content
   phenotypic readouts with neurons derived from human differentiated iPSCs with
   familial AD mutations, we will perform CRISPR based rescue screen for the
   various phenotypes associated with the mutations, such as endolysosomal
   transport, synaptic dysfunction, neuronal toxicity. The multiple-phenotypic
   rescue approach will enable the identification of novel key pathways and/or
   targets that could serve as drug candidates for the treatment of AD.

AUTOMATING CHEMISTRY IN THE AGE OF AI

Session Chair: Alex Godfrey, Ph.D., National Center for Advancing Translational
Sciences (NCATS) (USA)

 * Modern Automated Chemical Synthesis and Purification of Small Molecules
   Gerard Rosse, Arrival Discovery LLC
   
   Today, drug discovery remains a game of big numbers and many organizations
   routinely investigate small molecules collection in the 200,000 to 2 million
   compound range. Automation of chemical synthesis has gained a renewed
   interest to produce novel compounds and to facilitate the challenging
   multidimensional problem of compound optimization. This presentation will
   describe the strategy and obstacles to implement a technology platform for
   the production of 90,000 compounds per year in 13 mg quantity and >90%
   purity. Custom designed robotic instruments, specialized laboratory
   infrastructure, workflows, logistics and data management to enable high
   throughput synthesis and purification will be discussed. The implementation
   of core supercritical fluid chromatography (SFC) technologies provided a
   unique opportunity to increase productivity and significantly reduce
   operational costs. The presentation will conclude with an overview of the
   integration of chemistry automation with advanced compounds management
   systems.

 * A Beginner’s Guide to the Practicalities of Automating Chemical Synthesis
   Paul Harper, AstraZeneca
   
   In late 2017 AstraZeneca undertook an internal “hack-a-thon”, bringing
   together a diverse skill set to investigate our ambition of fully automated
   chemical synthesis for drug-like compounds. Over the past 2 years, we have
   evolved 4 prototypes to better understand the challenges associated with all
   stages of a multi-step synthesis process. In this presentation, we will
   review our most recent evolution, realizing fully automated batch and flow
   chemistries to fuel automated synthesis. We will describe in detail the
   integration and optimization of the Zinsser SOPHAS platform for batch
   chemical synthesis; along with the varied Waters devices for product
   purification and analysis.
   
   To achieve seamless integration, we have selected a third-party process
   scheduling software. Here we will cover the development of new drivers,
   protocols and interfaces for chemistry system control, along with the unusual
   demand of tracking and scheduling both single vial and associated plate-based
   activities (and the interplay between the 2 formats).
   
   Looking forward to our 5th iteration, we’ll discuss our plan for pre-cursor
   storage, along with our concepts for how reactions could be constructed using
   the Zinsser REDI platform.

 * SynFini: An Automated Chemical Synthesis Platform
   Nathan Collins, SRI Biosciences, SRI International
   
   Exploration in organic chemistry is still inherently a manual process both in
   conducting reactions in the lab and reporting results in the written
   literature. Both are subject to the skill of the practitioner and the next
   chemist who attempts to reproduce their reported results. In an effort to
   improve the transferability and reproducibility of chemistry we have
   developed an automation platform named SynFini that automates the design,
   reaction screening and optimization (RSO), and production of target
   molecules. SynFini includes three core components. A computational tool,
   SynRoute, develops synthetic strategies for molecules of interest. Routes to
   target molecules are built by combining knowledge of known chemistry from
   reaction databases and new reactions predicted by machine learning. A high
   throughput RSO platform, SynJet, (max. throughput reaction / s) enabled by
   inkjet printing experimentally validates these strategies. Automated analysis
   of the RSO outputs in the predicted route allows for the preparation of a
   digital synthesis protocol that drives a benchtop multistep synthesizer,
   AutoSyn. AutoSyn is capable of performing solution-based synthesis routes at
   the milligram to gram scale. To assist medicinal chemists in drug discovery
   programs, artificial intelligence (AI) can be included to aid in the design,
   selection, and prioritization of compounds with desired properties, such as
   biological activity and ADMET properties, and can be interfaced with SynFini
   automated synthesis and testing for rapid turnaround. Examples of how each
   tool works independently and then how they fit together into a seamless
   automated solution for design, synthesis and testing of a variety of
   molecules are presented. How such automated processes are digitally captured
   and electronically transferrable for ultimate reproducibility are discussed.

 * A Decade and Journey of Lilly’s Discovery Automated Synthesis
   James Beck, Eli Lilly
   
   Lilly actively engages Automated Synthesis in it's Medicinal Chemistry
   portfolio of projects. This brief talk introduces the journey Lilly has been
   on with the Automated Synthesis Lab (ASL - Indianapolis) and now with the
   closed-loop and integrated automation capabilities residing within the Lilly
   Life Sciences Studio (L2S2 - San Diego). Along the way, Automated Synthesis
   has provided a foundation for other Lilly initiatives including the Proximal
   Lilly Collection (PLC, published), Idea-to-Data (ItoD, published) and
   ChemoPrint (in press).

SCREENING AUTOMATION: MODULAR VS. HIGHLY INTEGRATED SYSTEMS

Session Chair: Helen Plant, B.Sc., AstraZeneca (UK)

 * Enabling Nontraditional Screeners from a Centralized uHTS Core
   Mitchell Hull, Calibr at Scripps Research
   
   Large, ultra-high-throughput screening systems can lead to an overreliance on
   simple assays and deny screening access to those with lower throughput needs.
   There is a temptation to forego slower, complex assays in favor of ones more
   amenable to HTS and to pursue familiar target families that “plug-in” to know
   platforms. HTS facilities, sometimes siloed within a department, are often
   unavailable even to those working in an HTS capable organization. As a
   result, some organizations have turned to multiple smaller systems designed
   for small-scale screening. However, data and compound management issues arise
   in this decentralized approach. Compound spotted assay plates, created by
   acoustic compound transfer platforms, can mitigate these issues. However,
   even combined, these cannot match the capabilities of a larger system when a
   larger campaign is needed. We have set up a hybrid uHTS/modular acoustic
   transfer platform that can act as one integrated system or three modular
   systems. Combining this with a high-value chemical library and an active
   pursuit of partners with high-value bioassays, we have pursued an approach to
   enable high-value, low-throughput assays, while maintaining uHTS capability
   and centralized compound and data management.

 * A B Cell Antibody Discovery Platform Using an ‘Islands of Automation’
   Approach
   Paul Anderson, Eli Lilly and Company
   
   In recent years Lilly has implemented a Next-Generation Research (NGR)
   initiative to improve the value output of the R&D portfolio. One of the NGR
   pillars focuses on decreasing the timeline to bring medicines to patients. As
   part of this initiative, Lilly has invested in an ‘Islands of Automation’
   approach to advance the B cell antibody discovery platform at the Lilly
   Biotechnology Center in San Diego to significantly increase throughput and
   reduce project timelines. This talk will focus on the automated systems that
   have been developed as part of this platform beginning with B cells sorted
   into microtiter plates through binding and functional assays run in
   dose-response plates from recombinantly expressed material. Examples of
   alternative instruments to traditional methods and software solutions to
   eliminate inefficiencies will be shared. This highly automated approach to
   antibody discovery has allowed us to meet aggressive timelines and
   dramatically increase throughput while allowing flexibility for future
   changes to our process.

 * Innovative Tube and Dispensing Technologies Enable Fully Acoustic Workflows
   for Drug Discovery Assays
   Silvio Di Castro, Sample Management / Discovery Sciences / AstraZeneca
   
   Innovative design and deployment of novel labware, instrumentation and
   software technologies have delivered an automated, fully acoustic platform
   and a step-change in small molecule Sample Management (SM) processes.
   
   For many years, conventional SM workflows have included multiple sample
   transfers between vessels, using a hybrid of contact and non-contact
   dispensing, which are cumulatively wasteful. These combine to affect
   excessive sample consumption, necessitating chemists to synthesize
   superfluous quantities of the compound.
   
   Here we show high-quality concordant datasets from the first fully acoustic
   workflow for physicochemical, enzymatic, cellular and in vitro ADME assays.
   We also show a reduction in (i) sample usage in these assays, (ii) DMSO usage
   throughout the process, and (iii) future synthesis requirements.
   
   An acoustically compatible storage tube (FluidX™ AcoustiX™ Sample Tubes,
   Brooks Life Sciences, UK) was designed with optimum geometry for dispensing
   accuracy and speed, whilst maintaining a working sample volume able to
   sustain a 10-year screening lifetime. Co-molded capping technology for these
   tubes has resulted in increased durability for multiple dispense access and
   sample longevity, whilst a novel split barcode at the base affords a central
   opening for transmission of the acoustic pulse.
   
   A tube-compatible acoustic liquid handler (Echo® 655T Liquid Handler, Beckman
   Coulter Life Sciences, USA) has been designed to utilize acoustically
   compatible storage tubes including a faster drop-transfer rate via a new
   transducer-focussing mechanism. A new dryer system removes moisture on the
   exterior of the tube, alongside local humidity control in the drop-transfer
   zone to maintain sample integrity.
   
   The development of a new, fully acoustic workflow has minimized sample
   handling and waste, enabling miniaturization of assays and hence reducing the
   amounts of sample required for synthesis to support drug discovery projects.
   We have implemented and validated novel labware and instruments for a
   transformative and sustainable solution to many drug discovery issues
   applicable across the industry.

 * Highly Integrated Modular Systems – Mobile Robots Unlock the Best of Both
   Architectures
   David Dambman, Biosero, Inc
   
   Choosing the right system architecture for your automation can be
   challenging. Large, highly integrated systems provide advantages in terms of
   throughput and operation simplicity, but the system itself becomes a single
   point of failure and it can be difficult to maintain up-time as well as
   evolve the system as applications and technologies change. Modular systems
   provide greater flexibility, are easier to scale and adapt to changing needs,
   but require more human effort to operate. It can also be challenging to
   integrate the data from disparate modules and manage efficient utilization
   across the full workflow.
   
   Fortunately, advances in mobile robot AGV (Autonomous Ground Vehicle)
   technology, coupled with new scheduling and data management architectures can
   bridge the gap. These provide the means to fully integrate modular, manual or
   robotic workcells by scheduling and executing operations with the additional
   capability to transport consumables, samples, and reagents between modules.
   This enables a truly connected and fully automated lab while still
   maintaining the advantages of standalone walk-up operations that has the
   flexibility to evolve as your needs change.

1536 AND BEYOND: HT MINIATURIZATION WHILE MAINTAINING PHYSIOLOGICAL RELEVANCE

Session Chair: Mindy Davis, Ph.D., National Institute of Allergy and Infectious
Diseases (NIAID) (USA)

 * Continued Development of High-Throughput MS and Applications for Cell
   Analysis
   Jonathan Wingfield, AstraZeneca
   
   Over recent years, AstraZeneca has worked to develop a high-throughput mass
   spectrometry platform that utilizes the speed and contactless nature of
   acoustics as a sample introduction technology. Fully automated acoustic mist
   ionization mass spectrometry platforms are now routinely used to support
   biochemical HTS campaigns, to date over 10 million samples have been
   successfully screened against more than 10 different enzyme targets using
   this technology.
   
   Having established a primary role for AMI-MS we are now looking to expand the
   application space where the technology could add value to early drug
   discovery. We have recently started to evaluate the impact of AMI-MS for
   metabolomic analysis of cell lysates, primarily within the early toxicology
   screening area.
   
   In December 2018, AstraZeneca and collaborators at several Swedish academic
   institutions and SME’s were awarded a phase 2 grant from Sweden’s Innovation
   Agency, Vinnova. The collaboration aims to develop technologies and workflows
   to enable primary patient-derived disease cells to be utilized in the early
   phase of drug discovery.
   
   This presentation will focus on the continued development of AMI-MS within AZ
   and how we are applying high-throughput mass spectrometry to enable clinical
   samples to be assessed in the early phases of drug discovery.

 * Array and Microfluidic-Based Cellular Assays Miniaturized Beyond the 1536
   Well Plate
   Jeffrey Gross
   
   Small molecule high throughput screening (HTS) in drug discovery
   traditionally involves microtiter plate screening in 384- and 1536- well
   formats. While these methods are miniaturized compared to Petri dishes or
   flasks, reagent and labor costs are still significant factors in high
   throughput screening campaigns. Here we present the development of cellular
   assays utilizing ultra miniaturized array-based and microfluidic devices. The
   experiments were aimed at reducing cellular assay volumes from uL to nL
   volumes. Challenges included maintaining environmental controls for cell
   health and handling small volumes for cells and compounds. Novel approaches
   in equipment, device design, and automation were required. Data suggest that
   cell health, cell morphology, and pharmacological responses to drugs were
   similar in nL volumes compared to those observed in 50 uL volumes in 384 well
   plates. The development of processes and automation to industrialize new
   devices will ultimately enable these technologies to be applied broadly in
   drug discovery. Cost savings in cell and reagent usage and the ability to use
   disease-relevant cell systems are paths toward reduced attrition in drug
   discovery.

 * Approaches that Enable Large-Scale Chemical Biology Interrogations
   Fred King, GNF Systems
   
   During SLAS (Lab Automation) 2009 we presented a low cost,
   automation-friendly, screening platform that used reporter gene assays
   (RGA’s) to delineate the interaction of small molecules with canonical
   mammalian signal transduction pathways. Our experience with this approach
   over the last decade has demonstrated its broad utility in the support of
   phenotypic screening, ranging from generating mechanism of action hypotheses
   for individual compounds to characterizing compound libraries. Furthermore,
   this RGA panel inspired and guided the development of additional platforms
   that are more comprehensive and flexible in terms of both cell types under
   investigation and the scope of biological activity detected. This
   presentation will focus on several of these new technologies, which all
   leverage Next Generation Sequencing technologies to measure RNA expression
   levels in a multiplexed fashion. The suite of approaches provides users with
   the ability to balance sequencing depth, transcriptome coverage and cost per
   well in their assay design. Coupled with internally designed automation
   platforms these systems allow expression levels of thousands of genes to be
   monitored in every well of an HTS-sized screen.

 * Identification of Chemical Compounds Inhibiting Zika Virus Replication
   Through a Large-Scale High-Content Screening Approach
   Laura Riva, Sanford Burnham Prebys Medical Discovery
   
   Zika virus (ZIKV) is a human mosquito-borne positive-sense RNA virus,
   belonging to the Flaviviridae family. World Health Organization (WHO)
   classified this virus as an Emergency in 2016 and currently identifies Zika
   as a priority disease. Although symptoms are generally mild, a risk of
   neurologic complications including Guillain Barré Syndrome is associated with
   the infection in adults, while infection during pregnancy is responsible for
   microcephaly and other congenital malformations. Since no vaccine or
   commercialized antiviral targeting this virus are available, scientific
   efforts are currently focusing on the development of treatments allowing to
   efficiently limit ZIKV spread. Prompted by this unmet medical need, we
   conducted a screen of 51,520 small chemical compounds using a high-content
   imaging cell-based assay, monitoring Zika virus replication within Huh-7.5
   cells by combining DAPI staining of cellular nuclei together with
   immunostaining of the Zika virus envelope protein. 99 candidates were
   identified and validated as inhibiting ZIKV replication of at least 50% at a
   concentration of 10 µM. Subsequent dose-response studies were performed to
   evaluate the effects of each compound on both virus replication and
   cytotoxicity and compounds showing a strong dose-response inhibitory effect
   on replication with weak cell toxicity were then selected for follow-up
   studies. Two compounds sharing a common structure presented a particularly
   promising antiviral activity with a selectivity index, calculated as the
   ratio of 50 % inhibitory (IC50) and 50 % viability (CC50) concentrations,
   greater than 30. This common chemical scaffold showed to specifically inhibit
   ZIKV, displaying an antiviral activity against several strains of both
   African and Asian lineages but no effect on other Flaviviruses tested. Its
   antiviral activity was confirmed with similar efficacy in more relevant
   models for ZIKV infection, including human monocyte-derived dendritic cells
   (hMDDCs), human neural progenitor cells (hNPC) and the placenta-derived
   choriocarcinoma cell line JEG-3. Time of addition kinetics as well as
   specific entry and replication assays excluded an inhibitory role during ZIKV
   entry, highlighting an antiviral role during the RNA replication step. This
   observation, in addition to the appearance of resistant mutant viruses upon
   selection in the presence of the drug, strongly suggested a non-structural
   protein of the virus as a target of the compound. Current efforts are ongoing
   to identify the specific viral target of the compound and to get more
   insights about its mechanism of action. In addition, Pharmacokinetics (PK)
   and in vivo efficacy studies will be performed in the near future to evaluate
   the therapeutic potential of this compound. In summary, taking advantage of a
   cell-based large-scale high-content screening approach to identify small
   chemical compounds showing antiviral activity against ZIKV, we identified a
   chemical scaffold specifically targeting this Flavivirus, inhibiting its RNA
   replication step.

OPEN SOURCE AUTOMATION

Session Chair: Sam Michael, National Center for Advancing Translational Sciences
(NCATS) (USA)

 * Improving Daily Operation of a Fully-Automated uHTS System
   Steven van Helden, Pivot Park Screening Centre
   
   Pivot Park Screening Centre (PPSC) is a small company that specializes in
   ultra-high-throughput screening (uHTS) for drug discovery. We perform about
   25 full deck screening campaigns a year on our own library of 300.000
   compounds, the European Lead Factory library of 550.000 compounds and client
   libraries up to 1.000.000 compounds. In order to support this huge
   production, we have implemented efficient processes on a fully automated
   screening system consisting of 3 robot pods integrated with a wide variety of
   instruments.
   
   Even in a highly automated environment like our uHTS lab, there is a
   continuous need for tools and little tricks to support our daily work. These
   include software tools to run active picking from the online store, re-use of
   (washed) verification plates, a simple tool for drying compound plates,
   implementation of a weighing station in the robot for checking dispensing
   performance, etc. Also, we have implemented cleaning stations for our heavily
   used certus dispensers. Finally, we make extensive use of a 3D-printer to
   create all sorts of tools in the lab and to save costs by printing parts of
   instruments that need repair. This presentation will provide insight into the
   daily operations in a uHTS lab.

 * Leveraging Open Source Electronics for Rapid Development of Custom Laboratory
   Devices
   Pierre Baillargeon, Scripps Research 
   
   The Lead Identification team at Scripps Florida routinely leverages open
   source technologies to meet operational challenges and to provide custom
   engineering tools for lab use. Most recently, these tools include the
   development of a micro solenoid dispensing QC platform built around the Lee
   Company VHS series valve. This system was developed to address the unmet need
   of dispensing of 3D models such as spheroids and to assist with general QC of
   valves used in ongoing HTS efforts. This QC platform allows users to
   characterize & optimize the performance of VHS valves under a variety of
   conditions.
   
   This reconfigurable QC platform is built on an optical breadboard and is
   comprised of three main subsystems: electronics, optical train and motion
   control. The electronics subsystem allows users to easily control VHS series
   valve behavior using an Arduino microcontroller through a custom in-house
   designed Arduino shield. The optical train consists of an off the shelf USB
   camera combined with an in-house designed open-source illumination panel that
   allows imaging of individual droplets via the stroboscopic effect. An
   open-source X/Y motion control system further increases the utility of the
   platform by allowing automated dispensing into microplates. User control of
   the QC platform is provided via a custom web-based interface that
   communicates directly with the microcontroller and allows users to easily
   specify microplate dispense patterns by interacting with graphic
   representations of microplate wells.
   
   Also presented is the development of a custom Arduino-based syringe pump
   system intended for use alongside the Lee valve QC platform. This syringe
   pump system utilizes a Tecan Cavro pump and allows for real-time adjustment
   of pump parameters during mixing and dispensing operations which are critical
   for mixing and homogeneous distribution of the spheroids. The development of
   these platforms, lessons learned and results of initial testing are
   presented.

 * Intelligent Microscopes Using Open-Source Hardware for High-Throughput
   Laboratory Automation
   Pavan Chandra Konda, Duke University
   
   Traditional microscopes used for automated imaging and analysis sets one
   aback with tens of thousands if not hundreds of thousands of dollars. This
   limits the number of microscopes a lab can afford, hence limiting the number
   of parallel experiments that can be performed. We present a novel approach by
   combining low-cost, low-resolution microscopes with advanced computational
   imaging methods that can extract high-resolution image information in the
   post-processing. In addition, we implement novel machine learning methods to
   jointly optimize the automation task, e.g. cell segmentation, and the data
   acquisition process, e.g. illumination pattern, to capture fewer data without
   losing the performance of the automated task.
   
   Our initial prototype costing ~$150 employed a Raspberry Pi as the computer
   and a modified Raspberry Pi V2 camera as the low-resolution microscope. A
   low-cost 16x16 LED array developed for display is used to illuminate the
   sample and 3D printed parts are used for assembly. LEDs in the array are
   sequentially illuminated to capture 256 low-resolution images, where the
   high-resolution information is encoded within these low-resolution images
   using the aperture synthesis concepts. The captured 256 low-resolution images
   were combined to achieve 0.8µm resolution, for the first time in a low-cost
   setting, across 4 mm2 field-of-view. The phase of the object is also
   recovered in the process, making this suitable for imaging cell cultures
   without any need of staining. In the latest developments, we implemented a
   new machine learning model to multiplex the illumination to reduce the number
   of images captured to two, without any loss in performance for tasks such as
   cell segmentation or detecting malaria infection. This also reduces the image
   processing time and, exploiting the increasing computing performance on
   opensource hardware such as Raspberry Pi and Google’s Coral Edge TPU, we are
   currently working towards achieving real-time machine learning-based
   automation on our portable low-cost setup.
   
   The 3D printed design of our microscope can be easily modified to the
   specific requirements of a lab, e.g. imaging stress fiber reorientation in
   cells under mechanical stimuli requires a different setup compared to imaging
   cell confluency in a petri-dish. Our optics and algorithms still stay valid
   for all these different configurations and the required modifications in the
   3D printed designs are usually minor. This is not possible with commercial
   systems that are designed for a limited number of imaging applications.
   Combining the latest developments in machine learning makes our approach a
   powerful tool for laboratory automation and diagnostics in low-resource
   settings.

 * Beyond High-Content Screening: An Open Next Generation Image Analysis
   Platform
   Peter Bajcsy, NIST National Institute of Standards and Technology
   
   There is an increasing interest in discoveries from images acquired by
   high-throughput and high content microscopy imaging of multi-well plates with
   biological specimens under a variety of conditions. As multi-dimensional
   automated imaging increases its throughput to thousands of images per hour,
   the computational infrastructure for handling the images has become a major
   bottleneck. The bottleneck associated challenges arise due to big image data,
   complex phenomena to model, non-trivial computational scalability that
   leverages the advanced hardware and cutting-edge algorithms, and incompatible
   software tools that vary in the language they were written in, the platform
   they were written for and capabilities they were designed to execute.
   
   To address the above challenges, groups have developed software solutions
   based on client-server systems with modern web technologies on the
   client-side and a spectrum of databases, computational workflow engines, and
   communication protocols on the server-side to hide the infrastructure
   complexity. However, these solutions have not focused on the interoperability
   of imaging specific computational plugins and visual exploratory capabilities
   of such plugins over very large image collections.
   
   To address these inter-operability and visual exploration challenges, the
   National Institute of Standards and Technology (NIST) and the National
   Institutes of Health (NIH) - National Center for Advancing Translational
   Science (NCATS) have formed a close collaboration to develop an open-source
   platform for executing web-based image processing pipelines over very large
   image collections with interoperable plugins. The plugins developed by both
   institutes are based on software containers as standardized units for
   server-side deployment, as well as on dynamically created web user interfaces
   (UI) to enter parameters needed for the software execution and for advanced
   visual data explorations on the client-side. Each container packages code,
   with all its dependencies, and has an entry point for running the computation
   in any computing environment. Each UI description file contains metadata
   about the plugin-container and the computation parameters.
   
   We will demonstrate the utility of the platform with algorithmic plugins by
   analyzing 1536 well plates with three spectral channels and multiple fields
   of views (FOVs) per well for drug dose-response across an array of features.
   Typical visual data exploration is assisted by algorithmic tools for quality
   control, stitching of FOVs per well, segmentation, characterization of
   regions of interest, and scalable visualization using Deep Zoom, a toolkit
   for browser viewing of gigapixel 2D images. The data explorations are
   interactive either in a Deep Zoom viewer or in a Jupyter notebook while
   prototyping pipelines. More demanding computations are supported via batch
   processing and deep learning-based pipelines are designed for GPU execution.
   With the NIST and NIH NCATS combined efforts, researchers are enabled to
   discover quantitative insights from their imaging data and reuse
   computational tools developed by anyone following the web computational
   plugin conventions.

Back to Top


BIOLOGICS DISCOVERY

Track Chairs: Rob Howes, Ph.D., AstraZeneca (UK) and Janice Reichert, Ph.D., The
Antibody Society (USA)

ANTIBODY THERAPEUTICS DISCOVERY: FOCUS ON BISPECIFICS AND IMMUNE CHECKPOINT
MODULATORS

Session Chair: Janice Reichert, Ph.D., The Antibody Society/mAbs (USA)

 * Creating a Novel T-cell Engaging Bispecific Antibody Platform: Fine-Tuning
   Anti-Tumor Activity with Sequence-Based Discovery and Machine Learning
   Katherine Harris, Teneobio
   
   Using a unique sequence-based discovery approach along with proprietary
   transgenic rats, we have created a large collection of fully human anti-CD3
   antibodies with diverse T-cell agonist activities. Our novel discovery
   platform combines antibody repertoire deep sequencing, high-throughput gene
   assembly, and recombinant expression. Our approach generates a large
   diversity of sequence-defined antibodies that we characterized in
   high-throughput T-cell agonist assays. Using machine learning tools, we were
   able to rapidly establish sequence-activity relationships and identify key
   residues and variable region positions in the antibody repertoire that had
   desired agonist behavior. The CD3 antibodies identified by our platform show
   diverse in vitro T-cell activation profiles measured by CD69 upregulation,
   IL2, and IFNg production. We also generated human domain antibodies targeting
   a variety of tumor antigens that we combined with our unique CD3 antibodies
   to create bispecific molecules that mediate redirected T-cell killing of
   tumor cells. In one particular example, we have created a panel of aCD3:aBCMA
   bispecific antibodies for the treatment of multiple myeloma that stimulate
   different levels of T-cell activity. Using a multiple myeloma tumor cell line
   along with primary human PBMCs, we demonstrate a spectrum of in vitro tumor
   cell killing activity with varying levels of cytokine release using our
   bispecific molecules with diverse CD3 binding activities. In summary, we have
   created a T-cell engaging bispecific antibody platform with tuned T-cell
   agonism that can be used to optimize the therapeutic index for a variety of
   tumor antigens.

 * Benefits of Chicken-Derived Antibodies for Combination Immunotherapy
   Torben Gjetting, Symphogen
   
   The development of novel antibodies and more powerful therapeutic
   combinations for immunotherapy is an intense area of focus. However, for
   difficult and/or conserved targets, finding antibodies with unique
   functionality, and generating early proof of concept pose challenges to the
   development of novel antibody therapeutics. Symphogen’s approach to discovery
   and development of potent antibody combinations for cancer immunotherapy
   using different species, including chicken, will be presented. Examples from
   our clinical pipeline will be shown.

 * Quantitative High-Throughput Screening Assays for the Discovery and
   Development of SIRPα-CD47 Interaction Inhibitors
   Thomas Miller, Institut Paoli Calmettes
   
   CD47 is an immune checkpoint molecule that downregulates key aspects of both
   the innate and adaptive anti-tumor immune response via its counter receptor
   SIRPα, and it is expressed at high levels in a wide variety of tumor types.
   This has led to the development of biologics that inhibit SIRPα engagement
   including humanized CD47 antibodies and a soluble SIRPα decoy receptor that
   are currently undergoing clinical trials. Unfortunately, toxicological
   issues, including anemia related to on-target mechanisms, are barriers to
   their clinical advancement. Another potential issue with large biologics that
   bind CD47 is the perturbation of CD47 signaling through its high-affinity
   interaction with the matricellular protein thrombospondin-1 (TSP-1). One
   approach to avoid these shortcomings is to identify and develop small
   molecule molecular probes and pretherapeutic agents that would (1)
   selectively target SIRPα or TSP-1 interactions with CD47, (2) provide a route
   to optimize pharmacokinetics, reduce on-target toxicity and maximize tissue
   penetration, and (3) provide for more flexible routes of administration. As
   the first step toward this goal, we report the development of an automated
   quantitative high throughput screening (qHTS) assay platform capable of
   screening large diverse drug-like chemical libraries to discover novel small
   molecules that inhibit CD47-SIRPα interaction. Using time-resolved
   fluorescent resonance energy transfer (TR-FRET) and bead-based luminescent
   oxygen channeling assay formats (AlphaScreen), we developed biochemical
   assays, optimized their performance, and individually tested them in
   small-molecule library screening. Based on performance and low false-positive
   rate, the LANCE TR-FRET assay was employed in a ~90,000 compound library
   qHTS, while the AlphaScreen oxygen channeling assay served as a
   cross-validation orthogonal assay for follow-up characterization. With this
   multi-assay strategy, we successfully eliminated compounds that interfered
   with the assays and identified five compounds that inhibit the CD47-SIRPα
   interaction; these compounds will be further characterized and later
   disclosed. Importantly, our results validate the large library qHTS for
   antagonists of CD47-SIRPα interaction and suggest broad applicability of this
   approach to screen chemical libraries for other protein-protein interaction
   modulators.

 * Discovery of High-Affinity, Pan-Allelic, and Pan-Mammalian Reactive
   Antibodies Against the Myeloid Checkpoint Receptor SIRPα
   Janet Sim, ALX Oncology
   
   Targeting the CD47-signal-regulatory protein α (SIRPα) pathway represents a
   novel therapeutic approach to enhance anti-cancer immunity by promoting both
   innate and adaptive immune responses. Ongoing clinical trials to inhibit this
   pathway by targeting CD47 has shown promising results in reducing tumor
   burden (Chow et al., ASCO 2019). Unlike CD47 which is expressed ubiquitously,
   SIRPα expression is mainly restricted to myeloid cells and neurons.
   Therefore, compared to CD47-targeted therapies, targeting SIRPα may result in
   differential safety and efficacy profiles, potentially enabling lower
   effective doses and improved pharmacokinetics and pharmacodynamics. In this
   talk, we will present our strategies of using wildtype/human antibody
   transgenic chickens for immunization and describe our screening approaches to
   identify SIRPα antibodies suitable for clinical translation. A total of 200
   antibodies were isolated and approximately 70 antibodies with diverse SIRPα
   binding profiles, sequence families, and epitopes were characterized. A
   subset of antibodies was shown to bind both human SIRPα v1 and v2 alleles
   with high affinity (nM-pM), potently antagonize the CD47/SIRPα interaction
   and potentiate antibody-dependent cellular phagocytosis in vitro. The
   anti-SIRPα antibodies also enhanced anti-tumor activity in both xenograft and
   syngeneic tumor models and were well tolerated in cynomolgus monkeys with
   favorable PK and extended receptor occupancy. These properties provide an
   attractive rationale to advance the development of these anti-SIRPα
   antibodies as a novel therapy for advanced malignancies.

CELL THERAPIES FOR CANCER: CAR-TS IN DEVELOPMENT

Session Chair: David Gilham, Ph.D., Celyad (BEL)

 * Generation of a Controllable CAR T Cell Therapy
   Travis Young, Calibr, a Division of Scripps Research
   
   T cell-based therapies, including T cell engaging bispecific antibodies, and
   genetically engineered chimeric antigen receptor engineered T cells (CAR-T
   cells) have produced remarkable results in clinical trials – achieving
   complete remissions in patients with hematological malignancies who failed
   multiple lines of prior therapy. Towards increasing the potency and safety of
   these therapeutics, as well as expanding them to cancers outside of the
   hematological space, we have defined how the biophysical characteristics of
   the antibody-based components of these therapies modulate the physiological
   response of the T cells that carry out the anti-tumor activity. For example,
   we have designed a “switchable” CAR-T cell system using antibody-based
   molecular switches. This platform enables fully tunable control of CAR-T cell
   activity in a universal format that can be redirected to nearly any
   therapeutic antigen target. The platform is expected to reduce the risk of
   severe adverse events that have plagued the development of CAR-T cell
   therapies clinically. We have demonstrated such a platform can reduce risks
   related to cytokine release syndrome and double as a safety switch to turn
   the therapy off in the case of an adverse event. We have further demonstrated
   the temporal control over CAR-T cell activation enables the development of
   robust central memory T cells. In preclinical mouse models, we’ve
   demonstrated these central memory cells can be recalled affording on-demand,
   in vivo T cell expansion. These concepts are expected to be central to
   clinical efficacy with T cell-based therapeutics and important to ultimately
   achieving efficacy in solid tumors. A proof of concept clinical trial will be
   initiated in late 2019 for patients with lymphoma.

 * Pluripotent Cell-Derived Engineered T and NK Cells as a Cornerstone Approach
   for Off-the-Shelf Cancer Immunotherapy
   Bob Valamehr, Fate Therapeutics
   
   Several obstacles currently hamper the broad use of adoptive cell therapies,
   including the inherent variability and cost of manufacturing of cellular
   populations, the absolute requirement for precise genetic editing of multiple
   elements and the elimination of undesired stochastic events associated with
   cellular engineering. Here we present a unique approach to create master
   pluripotent stem cell lines, clonally derived to contain precisely edited
   events at the single-cell level and the conversion of that master cell line
   into uniform populations of highly efficacious off-the-shelf engineered T and
   NK cells.

 * Exploiting Natural Killer Receptors for Autologous and Allogeneic CAR T Cell
   Therapy of Cancer
   David Gilham, Celyad
   
   Chimeric Antigen Receptor (CAR) T-cell therapy has hit the headlines with
   impressive clinical responses in hematologic B-cell malignancies that have
   led to the successful licensing of two products that both target CD19.
   Anti-BCMA CAR T-cell therapies for myeloma might come next but there is a
   dearth of targets outside of the B-cell malignancy space.
   
   Celyad has been exploring the potential of Natural Killer cell receptors to
   target cancer. Specifically, the company is conducting a series of clinical
   trials testing the safety and efficacy of CAR-T cells bearing the Natural
   Killer Group 2D (NKG2D) receptor that can specifically bind eight stressed
   induced ligands found on a broad range of cancers, yet largely absent from
   the surface of non-malignant, healthy cells.
   
   The first trial involved giving multiple infusions of autologous NKG2D-based
   CAR-T cells without pre-conditioning chemotherapy and provided some initial
   evidence of clinical activity with a good safety profile. Further clinical
   activity has tested this CAR-T approach with pre-conditioning and standard of
   care chemotherapy in hematological and solid tumors. Results of these trials
   and the company’s trial of allogeneic NKG2D-based CAR T cell therapy will be
   reviewed; the latter being thought to be the first allogeneic CAR-T approach
   to be tested in the solid cancers.
   
   Aside from NKG2D focused studies, the company is also embarking on a broader
   allogeneic CAR T cell platform technology exploiting interfering RNA to
   control graft versus host disease, the primary limitation of using allogeneic
   cells for therapy.
   
   Taken together, these early clinical studies suggest that the NKG2D receptor
   in both autologous and allogeneic approaches could provide the potential to
   target a broad range of tumor-targeting that could follow the success of CD19
   and BCMA CAR T cell therapy. The challenges ahead relate to how to exploit
   the targeting ability of NKG2D. Some of the next steps including manipulating
   the memory phenotype of the CAR T cell product will be discussed.

 * An Innovative Method for the Efficient, High-Throughput Transfection of
   Primary Human T-Cells
   Gregory Alberts, Lonza
   
   Cancer with 18.1 million new cases and 9.6 million cancer-related deaths
   observed in 2018 is still one of the most prevalent threats to human health
   and well-being. Therefore, there is a strong need for better cancer
   treatment. Cancer immunotherapy makes use of components of the immune system
   like antibodies that bind to, and inhibit the function of, proteins expressed
   by cancer cells. More promising novel immunotherapies rely on
   patient-derived, genetically modified cells like T-Cells or Natural Killer
   Cells that express chimeric antigen receptors (CAR).
   
   Primary Human T-Cells are difficult to modify genetically using chemical
   transfection reagents, just as virtually all non-dividing primary cell. Viral
   transduction methods depend on the cumbersome production of the viral
   vectors. Classical electroporation methods are often limited in throughput
   and can result in impaired cell viability and functionality. Therefore, we
   optimized the transfection and culture procedure for primary human T-Cells
   using the 4D-Nucleofector™ System and 96-well Shuttle™ Device allowing the
   high-throughput transfection of up to 96 independent transfection samples in
   parallel.
   
   Human T-Cells enriched from buffy coats were transfected with pmaxGFP™ Vector
   through a high viability or high efficiency Nucleofector™ program in 20 ?l
   volume. Donor-dependent transfection efficiencies of up to 70% with high cell
   viability were achieved 48 hours after transfection. Transfection of eGFP
   mRNA resulted in up to 60% transfection efficiency with more than 90% cell
   viability 24 hours after transfection.
   
   In a second step, we stimulated isolated human T-Cells for 2–3 days prior to
   transfection via CD3 and CD28. 1.0 x 106 cells were transfected with the high
   viability program using pmaxGFP™ Vector in 20 µl volume. Cells were analyzed
   24 hours post transfection revealing transfection efficiency and cell
   viability comparable to the results of unstimulated T-Cells.
   
   In a last evaluation step, using unstimulated human T-Cells, we could show
   very low intra- and interplate variability of the 96-well Shuttle™ System.
   Transfection efficiencies varied between 62% and 77%, while a cell viability
   of more than 80% compared to non-program control was observed.
   
   In summary, we present an efficient and reliable transfection system for
   primary human T-Cells that allows the parallel processing of up to 96
   independent samples. The showcased method will support cell-engineering
   approaches including screening of siRNA libraries, CRISPR-based genome
   editing and rapid evaluation of different CAR constructs to advance novel
   biomedical treatments including immunotherapy approaches.

FUTURE TECHNOLOGIES FOR BIOLOGICS DRUG DISCOVERY

Session Chair: Rob Howes, Ph.D., AstraZeneca (UK)

 * Target and Drug Discovery in ‘Undruggable Space’ Using Functional Proteomics
   Markus Muellner, PhoreMost
   
   Despite an increasing spend on drug development, many diseases remain
   unaddressed with little hope of finding new treatment options by conventional
   means. One reason for this is the lack of knowledge regarding which proteins
   are druggable, and which pockets on the protein surface might be most
   beneficially targeted by small molecules. Current methods for discovering new
   drug targets rely on genetic knock-out (CRISPR) or knock-down (RNAi) methods.
   While these techniques can be useful in providing candidate therapeutic
   target genes, the next step of developing protein-targeting therapies often
   stalls due to insufficient information on druggability. To address this
   problem, PhoreMost has developed a functional proteomics / phenotypic
   screening technology called “Protein Interference” (PROTEINi®) that yields
   both the target’s identity as well as information on available druggable
   sites within the target. PROTEINi utilizes proprietary large ( >1 Million),
   diverse, lenti-encoded libraries of small, self-folding, three-dimensional
   peptide "shapes", which are expressed in live cells and, much like small
   molecules, interfere and engage with available pockets on target proteins on
   a proteome-wide scale. In contrast to shRNA or CRISPR screens, PROTEINi works
   on the same level as most small molecules (the proteome) and is not
   influenced by gene copy number, SNPs or genetic buffering. The process
   discovers novel targets for a given assay system as well as peptides engaging
   this target functionally as a starting point for drug discovery. PhoreMost
   currently has internal small molecule programs in Oncology, Immuno-Onc and
   Neurodegenerative disorders. We have recently also expanded the method into
   Targeted Protein Degradation space to systematically discover novel and
   functional E3 linkage sites across a set of 600 ligases.

 * New Modalities for Drug Discovery
   Rob Howes, AstraZeneca
   
   Successfully drugging a target of interest is one of the key issues in drug
   discovery. Small molecules and antibodies have a long and successful history
   in drug development as our primary drug modalities. However, there are an
   increasing number of targets that are not amenable to these drug modalities –
   the ‘Undruggable Genome’. At AstraZeneca we are investigating new modalities
   to be able to drug this previously undruggable set of targets. In this talk I
   will describe our work with a range of new modalities including
   oligonucleoties, therapeutic proteins, cyclic peptides, antibody mimetics and
   PROTACS and how these are allowing us to tackle the ‘Undruggable Genome’.

 * µ-Hydroporator: A Next-Generation Intracellular Delivery Platform
   Aram Chung, Korea University
   
   The introduction of biomolecules and functional nanomaterials into cells is a
   crucial task in diverse biological situations, including immunotherapy,
   genome editing, regenerative medicine, and fundamental biological studies.
   Traditionally, intracellular delivery is achieved by carrier-based or
   membrane-disruption-based techniques. Carrier-based approaches utilize
   reconstituted viruses (e.g., lentivirus or AAV), or liposome (e.g.,
   Lipofectamine), and when optimized they offer effective delivery (e.g., DNA
   delivery for cell transfection). However, carrier-based approaches critically
   suffer from toxicity, low-throughput, and require time-consuming and/or
   labor-intensive preparation steps. Alternatively, membrane-disruption-based
   methods such as electroporation and microinjection create transient
   discontinuities on the cell membrane for target material diffusion. The
   physical cell membrane disruption is relatively independent of target and
   cell type, but they cause excessive damage to cells and suffer from limited
   throughput. To address these drawbacks, recent advancements in microfluidics
   and nanotechnologies have provided new solutions; however, identifying an
   ideal method that offers easy, low-cost, highly efficient, high-throughput,
   noninvasive and cell type/target independent delivery, remains challenging.
   Here, we present a next-generation intracellular delivery platform termed
   “µ-Hydroporator,” which introduces macromolecules into any cell type, at
   high-throughput, in a single-step, without a vector or external apparatus.
   µ-Hydroporator is purely based on the hydrodynamic cell
   deformation-restoration process, which opens the cell membrane and enables
   efficient transport of external target biomolecules or functional
   nanomaterials into the cell. In brief, the cell suspension mixed with target
   materials is injected into a T-junction microchannel with a micro-cavity
   where inertial vortices instantaneously deform cell. This rapid hydrodynamic
   cell deformation creates transient nanopores on the cell membrane, allowing
   the convective transport of foreign target molecules during the cell
   restoration process. Using µ-Hydroporator, we have successfully delivered
   diverse macromolecules (e.g., RNAs, Plasmids, DNAs, DNA origami,
   CRISPR-Cas9s, proteins, Q-dots, AuNPs, etc.) into various cell lines
   including difficult-to-transfect primary cell lines such as stem and immune
   cells, achieving highly efficient intracellular delivery (< 98%) in a
   high-throughput manner (~1,600,000 cells/min) while maintaining high cell
   viability (< 95%). Unlike traditional methods that rely on external
   apparatus, and/or chemical modification of target molecules, µ-Hydroporator
   only requires a syringe pump (not even a microscope!). This permits easy,
   robust and simple operation and cost-reduction from not requiring a skilled
   technician and instrument. We firmly believe that the reported µ-Hydroporator
   will establish a new paradigm in intracellular delivery, which will immensely
   benefit cellular engineering research and industry.

 * High-Throughput Encapsulation and Selection of Cells Based on Antibody
   Secretion Using Lab-on-a-Particle Technology
   Joseph de Rutte, University of California, Los Angeles
   
   We introduce a new approach to collect and quantify single-cell secretions
   without crosstalk in monodisperse droplets formed by precisely structured
   microparticles, enabling high-throughput screening based on this critical
   cell function. The ability to analyze and sort cells based on secretions
   (antibodies, cytokines, proteases, or other enzymes) has implications in
   understanding cellular heterogeneity fundamental to biology and creating new
   biotechnology products, such as biologics and cell therapies. Recently,
   droplet microfluidics has emerged as a powerful approach to perform
   single-cell secretion screening in high-throughput, using
   compartmentalization in a small volume to accumulate secreted factors to high
   levels for accurate detection. Despite this utility, the necessity of
   specialized equipment and expertise on the end-user hinders its widespread
   adoption. A platform that is fully compatible with standard lab equipment
   (e.g. pipettes, flow cytometers) has the potential to dramatically extend the
   reach of single-cell screening technology. Our particle-templated droplet,
   i.e. “Dropicle”, approach is unique in that pre-fabricated particles are used
   to form monodisperse emulsions that encapsulate single cells, requiring only
   standard lab equipment for the end-user. Cavity-containing microparticles are
   loaded into well plates and due to their morphology settle upright with their
   cavities exposed. Cells are loaded into the microparticle cavities and adhere
   via integrin binding sites. Biocompatible oil and surfactant are added and
   the suspension is agitated by pipetting to create incrementally smaller
   water-in-oil droplets. These resulting dropicles are monodisperse,
   maintaining a size defined by the particle geometry (CV < 6%), while the
   excess fluid is partitioned into surrounding smaller satellite droplets.
   Secretions from encapsulated cells are captured on the associated particles
   via protein A binding sites. Particles and associated cells and secretions
   are transferred back to the aqueous phase enabling downstream labeling and
   screening with standard flow cytometers. It was observed that seeded cells
   filled the cavities of the particles according to single-poisson statistics
   (in contrast to typical double-poisson statistics for single-cell,
   single-particle pairs in drops). After dropicle formation cells maintained
   high viability over 24 hours ( >80%). Initial tests with anti-IL-8 producing
   CHO cells demonstrate the ability to capture and label secretions on
   particles containing cells without crosstalk to neighboring particles.
   Further, we demonstrate the ability to isolate cells associated with high
   anti-IL-8 signals in high-throughput using commercial flow cytometry systems
   ( >100 sorts/s). Using this dropicle platform, researchers can perform
   droplet-based assays using standard lab equipment without sacrificing the
   precision of droplet microfluidics. Since dropicles are formed
   simultaneously, compartmentalization is rapid ( >400k in 30s) and can be
   easily scaled to accommodate large population screens. Further, the
   associated particle enables additional functionality such as physicochemical
   cues or cell-specific capture antibodies to select out specific cell types.
   Our results demonstrate new capabilities for lab-on-a-particle technologies
   that can accelerate the automation of single-cell assays.

Back to Top


CELLULAR TECHNOLOGIES

Track Chairs: Jason Ekert, Ph.D., MBA, GlaxoSmithKline (USA) and Nancy
Allbritton, M.D., Ph.D., University of North Carolina Chapel Hill (USA)

DEMONSTRATING TRANSLATIONAL RELEVANCE OF ORGANOID AND MULTICELLULAR TISSUES FOR
PRECLINICAL MODELS OF HUMAN DISEASE

Session Chair: Nancy Allbritton, M.D., Ph.D., University of North Carolina
Chapel Hill (USA)

 * Advanced Bioprinting Strategies for Tissue and Tissue Model Fabrication
   Y. Shrike Zhang, Harvard Medical School
   
   Bioprinting has recently emerged as an enabling technology in tissue
   biofabrication at high fidelity. This talk will discuss our recent efforts on
   developing a series of advanced bioprinting strategies, including sacrificial
   bioprinting that allows generation of perfusable microchannels embedded in
   hydrogel microchannels, microfluidic and hollow fiber bioprinting that
   achieves production of standalone cannular tissues, and multi-material
   bioprinting based on both extrusion and stereolithographic modalities that
   enables creation of complex hierarchical tissue microstructures. Innovations
   in various cytocompatible and bioactive bioink formulations will also be
   presented. These platform bioprinting methods have been demonstrated to
   facilitate faithful fabrication of biomimetic tissues and their models
   spanning from the heart, liver, and musculoskeletal system to blood vessels
   and beyond, as well as their diseased forms, for applications in regenerative
   medicine and inaccurate screening of therapeutic agents.

 * Wound-Conformal Delivery of Dermal Tissue Constructs for Full-Thickness Burn
   Treatment Using a Handheld Bioprinter
   Richard Cheng, University of Toronto
   
   Full-thickness burns where both the dermal and epidermal layers of the skin
   are destroyed result in high patient mortality due to infection, dehydration,
   and shock. The current standard of care involves the direct application of an
   acellular crosslinked protein scaffold which forms a temporary physical
   barrier and promotes host cell migration into the wound area; however, this
   is problematic in severe burns where little healthy skin is available for
   repair. Delivery of patient-derived autologous or immunoprivileged allogeneic
   cells are emerging as potential treatment options due to continuous
   extracellular matrix remodeling and persistent cell signaling, but challenges
   include homogenous delivery of cells onto a large, non-flat wound topography.
   Although approaches such as cell spraying and microparticle injecting have
   been explored in the field, the continuous formation of three-dimensional,
   hydrogel-based tissue constructs uniformly on a physiological wound surface
   remains unsolved. Here, we report the development of a handheld bioprinter
   that delivers wound-conformal dermal tissue constructs to improve wound
   healing in full-thickness burns. Mesenchymal stromal cell (MSC)-containing
   fibrinogen bioink and thrombin crosslinker solutions were delivered through
   on-board syringe pumps to a microfluidic printhead with internal bifurcated
   channels. Dermal tissue constructs of consistent thickness covered with the
   crosslinker were obtained at the exit. Wound-conformal delivery of these
   MSC-laden dermal tissue constructs was achieved by translating the printhead
   along the wound surface by a soft silicone wheel, while a two-axis gimbal
   design allowed it to adapt to the wound topology. We observed that the
   addition of 1% hyaluronic acid (HA) provided desirable shear-thinning
   behavior of the bioink (1.2 Pa•s at shear rate 1/s; 0.35 Pa•s at shear rate
   100/s), resulting in 83% of the starting thickness to be maintained for
   deposition surfaces with inclination angles of 45 degrees. Furthermore, these
   fibrin-HA hydrogels maintained high biocompatibility with the co-delivered
   MSCs ( >94%), in addition to the long-term preservation of 3D morphology and
   cell proliferation as shown with Hoechst/Phalloidin+ immunostaining over one
   week. To demonstrate the clinical utility of this approach, we uniformly
   distributed 1x10^6 MSCs/ml of the fibrin-HA hydrogel on a porcine 5cm x 5cm
   full-thickness burn wound model and quantified a 1.4-fold improvement of
   macroscopic re-epithelialization speed, a 1.3-fold increase in collagen
   density in the dermal layer, and a 2.5-fold reduction in CD11b+ inflammatory
   cell activity after 28 days compared to burn controls, as observed via
   microscopic analysis of H&E histological stains. Taken together, we have
   shown that the handheld bioprinter can conformally deliver MSC-containing
   dermal tissue constructs directly on wound substrates with physiological
   topographies, leading to full-thickness burn wound repair as shown in porcine
   pre-clinical case studies.

 * Generation, Validation and Application of Induced Pluripotent Stem Cell
   Models for Functional Genomics
   Lisa Mohamet, GSK
   
   Myeloid cells play critical roles in adaptive and innate immunity and
   dysregulation can result in disease pathology, such as neurodegeneration.
   However, a detailed mechanistic understanding of human myeloid biology has
   been hampered by the lack of robust and scalable models for cellular and
   genetic studies. Conventional approaches rely upon immortalized cells that
   lack biological relevance or primary cells which are limited in number,
   reproducibility, and genetic perturbation. To overcome these challenges, we
   developed and industrialized a human induced pluripotent stem cell
   (iPSC)-derived myeloid platform that permits a robust and continuous supply
   of progenitors that are subsequently differentiated into macrophage or
   microglia. Since each iPSC line retains the genetic information of the donor
   this provides an opportunity to harness human genetics to investigate in
   vitro disease mechanisms.
   
   We performed extensive transcriptomic, epigenetic, proteomic and metabolomic
   analyses with concomitant phenotypic (e.g. flow cytometry, image analysis)
   and functional assays (e.g. phagocytosis, cytokine secretion) to support
   their use as a model to primary counterparts. Here, we demonstrate a
   combination of conventional and innovative technologies to generate and
   validate iPSC-derived target cell types as an unlimited source of patient
   genotype-specific cells to study. We describe the implementation of such
   disease-relevant models to enable large scale (epi)genomic functional
   modeling for improved novel target ID.
   
   The human biological samples were sourced ethically and their research use
   was in accord with the terms of the informed consents under an IRB/EC
   approved protocol.

 * High-Throughput Organoid and Monolayer Platforms to Study Intestinal
   Physiology
   Scott Magness, University of North Carolina, Chapel Hill
   
   Intestinal organoid technologies have revolutionized culture models to study
   physiology, disease, and injury in vitro. While primary stem cell-driven
   organoid cultures offer many improvements over conventional cancer cell line
   models, individual organoids are highly heterogeneous in lineage ratios,
   morphologies, growth properties, and other physiological parameters.
   Additionally, the enclosed lumen prohibits easy access to the apical cell
   surface to study nutrient absorption, the microbiome, and drug interactions
   with the epithelium. We have developed platforms that address these
   challenges. Specifically, our group focuses on engineering high-throughput
   systems to study single-cell stem cell biology, stem cell niche co-cultures,
   organoid dynamics, luminal physiology, and the microbiome. These platforms
   can be applied to organoids across any tissue type, are scalable, portable,
   and represent a high-resolution and statistically robust solution for
   preclinical models of human disease.

APPLICATION OF ORGAN-ON-CHIPS AND MICRO-PHYSIOLOGICAL SYSTEMS

Session Chair: Deok-Ho Kim, Ph.D., Johns Hopkins University School of
Medicine (USA)

 * Modeling Dystrophic Cardiomyopathy on a Chip for Phenotypic Drug Screening
   Deok-Ho Kim, Johns Hopkins University School of Medicine
   
   Directed differentiation of human pluripotent stem cells (hPSCs) into
   cardiomyocytes typically produces cells with structural, functional and
   biochemical properties that most closely resemble those present in the fetal
   heart. Here we establish an in vitroengineered developmental cardiac niche to
   produce matured hPSC-derived cardiomyocytes (hPSC-CMs) with enhanced
   sarcomere development, electrophysiology, contractile function, mitochondrial
   capacity and a more mature transcriptome. When this developmental cardiac
   niche was applied to dystrophin mutant hPSC-CMs, a robust disease phenotype
   emerged, which was not observed in non-matured diseased hPSC-CMs. Matured
   dystrophin mutant hPSC-CMs exhibited a greater propensity for arrhythmia as
   measured via beat rate variability, most likely due to higher resting
   cytosolic calcium content. Using a custom nanopatterned microelectrode array
   platform to screen functional output in hPSC-CMs exposed to our engineered
   developmental cardiac niche, we identified calcium channel blocker,
   nitrendipine, mitigated hPSC-CM arrhythmogenic behavior and correctly
   identified sildenafil as a false positive. Taken together, we demonstrate our
   developmental cardiac niche platform enables robust hPSC-CM maturation
   allowing for more accurate disease modeling and predictive drug screening.

 * Engineering the Cell Niche to Enhance iPSC-derived Cardiomyocyte Maturity and
   Predictivity in a High-Throughput, Assay-Agnostic Manner
   Nicholas Geisse, NanoSurface Biomedical
   
   Stem cell technology holds great promise for mitigating the cost of drug
   development by properly modeling human biology in vitro. Assays based on
   these technologies have the potential to be more predictive of toxicity and
   efficacy when compared to simpler single-molecule assays. Building these
   representative organ-on-chip models relies on generating hierarchically
   organized cells and tissues. In vivo, this organization is driven by a
   complex interplay of cells and their environment, including the extracellular
   matrix (ECM). Traditional cell culture environments—typically composed of
   hard and unstructured glass or plastic— fail to fulfill the role of the ECM
   in development. Consequently, many in vitro stem cell models often fall short
   incorrectly reproducing critical in vivo phenotypes because cultured cells
   oftentimes lose type-specific characteristics or express phenotypes
   indicative of an immature developmental stage. Considerable effort is
   directed at fabricating biomimetic culture environments that maintain or
   promote mature phenotypes. However, making biomimetic substrates typically
   involves costly or hard-to-reproduce techniques that are often incompatible
   with many standard assays; these challenges are compounded when using
   high-throughput techniques. Our objective is to develop novel surfaces that
   mimic the mechanical and structural cues of the ECM without compromising
   compatibility with state-of-the-art assays and instruments. The fabrication
   scheme is based on high-precision photolithography techniques and is thus
   highly reproducible, scalable and amenable to integration with most
   industry-standard endpoint assays, including high-NA microscopy. Further, the
   approach is centered on SLAS/ANSI/SBS-compliant formats that are compatible
   with high-throughput automated platforms. Our data demonstrate that various
   cell types are amenable to this approach. hiPSC-derived cardiomyocytes (CMs)
   showed in vivo-like myofibril alignment, sarcomere spacing and width, and
   expression of CM-specific proteins that are present in mature myocytes.
   Higher-ordered 2D anisotropic myocyte tissues also showed adult-like
   structure and electrophysiological responses to drugs in vitro when compared
   to traditional unordered 2D isotropic constructs. Examples of phenotype
   enhancement of other adherent mammalian cell types will be presented, further
   demonstrating the utility of the approach for generating more representative
   cells and tissues. Finally, we extend this technique to pattern the surface
   of microelectrode arrays and demonstrate that these ECM-based cues enhance
   the electrophysiological response of cardiomyocytes to various drugs of known
   action. We conclude that our approach is a viable method for re-creating
   specific aspects of the ECM that are critical for driving the development and
   maturation of stem cells in culture.

 * Automation of Multi-Organ Chip Assays and Microscopic Analysis
   Ann-Kristin Muhsmann, Technische Universität Berlin
   
   Microphysiological systems (MPS) are designed to mimic human organs and
   physiology with the aim of improving the drug development process. They have
   proven to be a powerful tool at the research level and a solid basis for the
   establishment of qualified preclinical assays with improved predictive power.
   However, one remaining drawback is the lack of comprehensive standardization.
   This hinders their use for regulatory purposes as well as it impedes the
   extraction of the full extent of information from acquired data. Moreover,
   one main advantage of MPS in contrast to animal testing – the insight in “the
   body” throughout the test assays – cannot be deployed fully as long as assay
   execution, observation and analysis are highly time-consuming and resource
   binding. This is why there is an immediate demand to automate MPS cultivation
   and analysis. Concluding, we developed a dedicated system for the cultivation
   of our TissUse Multi-Organ-Chips.
   
   The Humimic AutoLab cultivates up to 24 Multi-Organ-Chips (MOCs)
   simultaneously, providing customized incubation and systemic pulsatile media
   circulation. Regular media exchanges, substance application and sample
   extraction are executed automatically according to assay specifications.
   Routine microscopic analyses such as bright field imaging and fluorescence
   measurements are conducted in scheduled cycles and at any chosen time. To
   hinder contamination and for operator protection, all liquid handling steps
   are conducted under sterile conditions. Media, substances and samples are
   stored in a refrigerator at 4°C, which are delivered on-demand via an
   automatic provisioning system. Cell culture material such as well plates and
   single-use pipet tips last for a minimum of four days until a restock is
   necessary. The refrigerator also holds media samples and stores them until
   further analysis. Dedicated software allows for a coherent and time-efficient
   input of all assay parameters as well as it provides a variety of tools for
   data analyzation. The Humimic LabOS also allows for assay feasibility checks
   and provides the operator with instructions for equipping the system. The
   fully automated image acquisition and ensuing analyzation will finally allow
   for higher comparability of results and new findings through pattern
   recognition and machine learning algorithms. Transferring our
   well-established co-culture assays with organ models such as liver, skin,
   intestine and bone marrow organoids proofed successful and showed high
   comparability to the manually conducted assays.

 * Novel Oxygen-Gradient Platform for the Co-Culture of Anaerobic Gut Microbiota
   with Primary Human Colon Epithelium
   Raehyun Kim, University of North Carolina at Chapel Hill and North Carolina
   State University
   
   Humans have co-evolved with their gut microbiota in a symbiotic relationship
   essential for health, yet how these thousands of bacterial species influence
   human biology remains little understood. A better understanding of the
   interplay between human cells and gut microbiota is required to exploit the
   complex relationships responsible for the local and distant effects of the
   microbiome on the human body. Accordingly, significant interest exists in the
   biotechnology community for improved in vitro models of the human
   gastrointestinal system, in particular models that support human-microbial
   co-culture. This feat is complicated by the fact that over 99% of gut
   bacteria are obligate anaerobes that die in 30-60 min after exposure to room
   air. Therefore, we have developed an easy-to-use and intuitive platform to
   replicate the steep O2 gradient across the in vivo colonic epithelium, thus
   create the appropriate environment required for anaerobes while maintaining
   viable, healthy epithelial tissue. We have computationally modeled, designed
   and prototyped the co-culture platform to fit within an SBS standard 12-well
   plate. The co-culture platform consisted of a basal reservoir and luminal
   reservoir with a porous polyester membrane and extracellular matrix (ECM)
   support dividing the two reservoirs. Using our culture methods, colonic
   epithelial stem cells were expanded on the ECM support and subsequently
   differentiated into all cell types found in the intestines in a monolayer
   ideal for compound screens and luminal stimulation/co-culture. Additionally,
   the ECM could be micro molded to recreate the physical architecture of the
   colon. Once the colonic epithelial layer was established, the luminal
   reservoir was sealed with an O2-impermeable barrier which resulted in the
   auto-generation of an anoxic environment (< 2% O2) in the luminal reservoir
   within 8 hours by the O2 consumption of the epithelial cells. The basal
   chamber remained normoxic to supply the epithelial cells with O2 through the
   porous membrane and ECM support. The generation of a steep O2 gradient was
   measured and experimentally confirmed. The resulting O2 gradient allowed for
   anaerobes (lactobacillus rhamnosus GG) to be cultured in the luminal
   reservoir in contact with an oxygenated colonic epithelial layer. Colonic
   epithelium and anaerobic bacteria each maintained >90% viability when
   co-cultured for ≥3 days. Our co-culture platform is simple, robust,
   self-sustaining and easy-to-use. It does not require any fluidic and gas
   control systems. It is based on a regular standard SBS microplate format that
   industry and academia use daily. Thus, it can be adopted in any microbiology
   laboratory without requiring new equipment.

DEVELOPMENT AND APPLICATIONS OF FUNCTIONAL GENOMICS TECHNOLOGIES

Session Chair: Alejandro Amador, Ph.D., GlaxoSmithKline (USA)

 * Application of Genome-Wide Arrayed CRISPRn Screening for Target Discovery
   Davide Gianni, AstraZeneca
   
   Identification of novel and translatable therapeutic targets is urgently
   required for diseases with unmet clinical needs. Application of CRISPR/Cas9
   technology in an arrayed screening format holds great potential for rapidly
   identifying new targets from physiologically relevant models. We have
   developed an end-to-end arrayed CRISPRn library screening platform for target
   discovery and integrated it with the capacity to interrogate advanced cell
   models. We are using this platform to perform gene perturbation experiments
   with the aim of identifying novel targets, understanding compound mode of
   action, building patient stratification hypotheses and ideas for novel
   combination therapies. A number of case studies will be presented
   highlighting the progress we have made in making this platform amenable to
   screening complex and advanced cell models. Taken together, these findings
   underline how functional genomics approaches using CRISPR/Cas9 technology are
   starting to revolutionize the drug discovery process.
   
   Identification of novel and translatable therapeutic targets is urgently
   required for diseases with unmet clinical needs. Application of CRISPR/Cas9
   technology in an arrayed screening format holds great potential for rapidly
   identifying new targets from physiologically relevant models. We have
   developed an end-to-end arrayed CRISPRn library screening platform for target
   discovery and integrated it with the capacity to interrogate advanced cell
   models. We are using this platform to perform gene perturbation experiments
   to identify novel targets, understanding compound mode of action, building
   patient stratification hypotheses and ideas for novel combination therapies.
   A number of case studies will be presented highlighting the progress we have
   made in making this platform amenable to screening complex and advanced cell
   models. Taken together, these findings underline how functional genomics
   approaches using CRISPR/Cas9 technology are starting to revolutionize the
   drug discovery process.

 * Leveraging Functional Genomics in Oncology: A High Throughput Biology
   Platform for Novel Drug Target Candidates
   Alejandro Amador
   
   New advances in gene editing, such as CRISPR-Cas9 technology, open new and
   exciting avenues for genome-scale functional interrogation of the genome. The
   newly formed Functional Genomics department (FxG) at GSK exists to provide
   the deep scientific knowledge and technical capabilities required to reveal
   genetic clues that underpin human disease, ultimately providing better
   medicines for our patients. Our High Throughput Biology and Imaging (HTBI)
   group in FxG has designed and built a high throughput automated robotic
   platform to interrogate drug and genetic interactions (DrugxDrug, DrugxGene
   and GenexGene) at scale by using cell-based high content imaging, flow
   cytometry and plate reader screening assays. The group is formed by a
   multidisciplinary team of scientists, including assay development and high
   content imaging experts, computational scientists and automation engineers.
   Our primary goal is to use functional genomics approaches for predicting and
   identifying new oncology combination therapies, and immuno-oncology
   mechanistic insights of GSK assets mode of action by interrogating gene
   function using RNAi and CRISPR/Cas9 genome wide array screening. The
   presentation will have examples on a couple of projects we are currently
   working on highlighting the potential benefits to run a functional genomics
   screening at scale.

 * Using CRISPR-Cas9 Screening to Identify Genes Modulating the
   Nigericin-Induced Pyroptosis
   Christian Parker, Novartis
   
   Inflammasomes are multiprotein complexes that sense danger or
   damage-associated molecular patterns, DAMPS, as part of the innate immune
   system. This recognition leads to the release of cytokines and other
   signaling molecules that can then lead to cell death. Typically the term
   inflammasome refers to the complex of proteins including PYCARD (ASC), NLRP3
   (NLRC4 or AIM2) and pro-caspase-1. Upon activation by various DAMPS this
   multi-protein complex promotes activation of caspase-1, which then leads to a
   cascade of events that cause the release of intercellular signals such as
   IL-β and IL-18. These danger signals, as well as released intracellular
   components, can then further activate inflammasomes present in surrounding
   cells. In addition, the activation of the inflammasome and caspase-1 can also
   lead to cell death due to the activation of membrane pores such as gasdermin
   D.
   
   Mutations in components of the inflammasome have demonstrated this as a key
   pathway regulating autoimmune diseases; e.g. cryopyrin-associated periodic
   syndrome (CAPS), pyrin-associated autoinflammation with neutrophilic
   dermatosis (PAAND) and Familia Mediterranean Fever (FMF). A detailed
   understanding of the constituents of the inflammasome, and the pathway
   leading to its activation will have utility in designing treatments for a
   range of diseases associated with inflammation.
   
   This report describes the development of an assay monitoring the induction of
   inflammasome mediated cell death (pyroptosis). The development of this assay
   allowed a genome-wide CRISPR-Cas9 screen to identify genes that regulate
   assembly and activation of the inflammasome. The assay utilized nigericin
   induction of the NLRP3 inflammasome mediated cell death in PMA differentiated
   THP-1 cells.
   
   The screen successfully identified known components of the inflammasome as
   well as several genes that have not been previously implicated in
   inflammasome induced cell death. The use of a genome-wide screen allowed a
   comprehensive evaluation of the pathways controlling inflammasome assembly
   and activation. The top 1000 genes were identified for the creation of a
   focused mini-pool library of potential targets. Retesting of this mini-pool
   of potential targets confirmed the activity of many of these genes as
   modulating the inflammasome. So a further selection of genes was then made
   and these genes were knocked out individually using CRISPR-Cas9.
   
   This presentation will discuss a number of the challenges faced invalidation
   of genes using this system as well as discussing potential means to address
   these issues.

 * Presentation Title TBD
   TBD

Back to Top


DATA ANALYSIS AND INFORMATICS

Track Chairs: Yohann Potier, Ph.D., Voyager Therapeutics (USA) and Nicola
Richmond, Ph.D. GlaxoSmithKline, (UK)

THE LAB OF THE FUTURE: AUTOMATION IN THE DIGITAL AGE

Session Chair: Umesh Katpally, Ph.D., Novartis Institutes for Biomedical
Research (USA)

 * The Lab of the Future: Automation in the Digital Age
   Michael Shanler, Gartner, Inc.
   
   "Lab of the Future" (LoF) has recently become a popular topic for modernizing
   laboratories. While performing upgrades to laboratory informatics systems
   such as ELN or LIMS and adopting "hyped" technologies such as IoT, AI/ML,
   AR/VR and blockchain may support modernization on the surface, many existing
   LoF strategies run the risks of only delivering incremental value. Your LoF
   strategies need to have a deeper impact to survive executive scrutiny and
   must put an augmented data analytics strategy at the center. A LoF strategy
   must also enable a digital twin for the lab at multiple levels- with impacts
   on lab assets, personnel, and systems. As businesses transform, all aspects
   of business, including the laboratory need to support digital optimization
   and transformation. In this session, we review the meaning of digitalization,
   the technologies important for achieving LoF and outline strategic steps for
   ensuring your "LoF" strategy will be aligned to deliver true value in the
   Digital Era.

 * How Advances in Mobile, Voice and AI Technology are Impacting Scientists: The
   Evolution of Technology in the Lab - University of California, San Francisco
   Case Study
   Ernesto Diaz Flores, University of California San Francisco
   
   Scientists working today must navigate very large and complex datasets and
   work within regulatory boundaries that are tighter than ever. To meet the
   needs of modern scientists, lab documentation and management systems have had
   to evolve from simple pen and paper to flexible, integrated digital tools.
   
   We are at an era in which technology is at our fingertips, and having new lab
   automation tools like a voice-powered AI digital lab assistant that allows
   integration of multiple functionalities within a laboratory annotation system
   greatly simplifies research workflows.
   
   Ernesto Diaz-Flores is an Assistant Adjunct Professor at UCSF who works with
   his team of scientists in the lab to study novel therapeutic targets for
   high-risk subtypes of childhood leukemia. New technologies such as
   voice-powered AI digital assistants enable scientists at the UCSF lab to take
   voice notes, upload photos of experiments in real-time, set up several
   reminders throughout the day and even dictate what reagents they need in
   their shopping list, and have it all immediately added to their e-lab
   notebooks. There’s also been less human error, scientists can capture and
   access more information at the point of experimentation hands-free. When eyes
   and hands are occupied on the experiment, their voice can make the
   observations and capture the information in real-time with digital lab
   assistants.
   
   As mobile, voice and AI technology evolve, there are now new options for
   scientists that seamlessly integrate with lab equipment and other data
   sources. The developments in mobile, voice and AI/machine learning technology
   are playing an important role in helping scientists bring their innovations
   and discoveries to market, improve efficiencies in the lab and make their
   work more reproducible.

 * How Advances in Mobile, Voice and AI Technology are Impacting Scientists: The
   Evolution of Technology in the Lab - University of California, San Francisco
   Case Study
   Gursatya "Guru" Singh, LabTwin
   
   Scientists working today must navigate very large and complex datasets and
   work within regulatory boundaries that are tighter than ever. In order to
   meet the needs of modern scientists, lab documentation and management systems
   have had to evolve from simple pen and paper to flexible, integrated digital
   tools.
   
   We are at an era in which technology is at our fingertips, and having new lab
   automation tools like a voice-powered AI digital lab assistant that allows
   integration of multiple functionalities within a laboratory annotation system
   greatly simplifies research workflows.
   
   Ernesto Diaz-Flores is an Assistant Adjunct Professor at UCSF who works with
   his team of scientists in the lab to study novel therapeutic targets for
   high-risk subtypes of childhood leukemia. New technologies such as
   voice-powered AI digital assistants enable scientists at the UCSF lab to take
   voice notes, upload photos of experiments in real-time, set up several
   reminders throughout the day and even dictate what reagents they need in
   their shopping list, and have it all immediately added to their e-lab
   notebooks. There’s also been less human error, scientists can capture and
   access more information at the point of experimentation hands-free. When eyes
   and hands are occupied on the experiment, their voice can make the
   observations and capture the information in real-time with digital lab
   assistants.
   
   As mobile, voice and AI technology evolve, there are now new options for
   scientists that seamlessly integrate with lab equipment and other data
   sources. The developments in mobile, voice and AI/machine learning technology
   are playing an important role in helping scientists bring their innovations
   and discoveries to market, improve efficiencies in the lab and make their
   work more reproducible.

 * The Hyperloop for the Lab: An Integrated Approach for Sample Delivery and
   Treatment of Culture Dishes
   Christoph Otto, TU Dresden
   
   Digitalization, Automation and Miniaturization currently change the way we
   live and work. It also affects the daily work in laboratories creating what
   we perceive as the Lab 4.0 or the Lab of the Future. The disruptive
   development of new technologies such as open-source automation technology,
   the Internet of Things (IoT) and 3D-printing offer endless possibilities for
   the rapid engineering of new laboratory devices, which are compact, adaptable
   and smart. In conjunction with automated 3D-image analysis or deep learning
   algorithms, powerful instruments emerge to create and resolve research data.
   At the SmartLab systems department, the PetriJet31X hyperloop technology was
   developed to automate all processes associated with culture dishes in
   environments such as routine laboratories or culture development for the next
   generation of antibiotics. The device technically is an x-y-robot consisting
   of two linear axles enabled to transport all kinds of culture dishes from A
   to B through a 3D-printed gripper-system which can also remove the lid of the
   culture dish. The platform has been extended with a rail system and a small
   robot with the ability to transport piles of culture dishes or other
   laboratory material throughout the lab. The core part of the programming is a
   self-learning control software that does not need any teaching – the most
   time-consuming part of setting up a typical robot. With the presented
   solution an experiment conducted on samples is planned only once and executed
   for all culture dishes in the machine with the right processing station
   installed – e. g. 3D sample imaging and analysis. It is no longer necessary
   to specify locations for culture dish piles and treated dishes get allocated
   dynamically and drawn e.g. from the incubating chamber while user
   interactions are directed by LED-lighting. The system can process more than
   1.200 culture dishes in an 8-hour shift and is equipped with a storage unit
   for these culture dishes. One example of the benefit of the PetriJet
   hyperloop can be found in routine labs for water and food inspection. Large
   numbers of samples get incubated on specific medium in culture dishes and are
   visually inspected regularly. Our system directly receives the tasks from the
   laboratory information and management system (LIMS), creates job lists and
   provides analytical data to the lab assistants through the LIMS. The data can
   then easily be turned into result sheets right from the desk. The unique
   feature of the system is that it can operate the night shift with no staff
   present. The PetriJet31X hyperloop platform now operates at the Chair of
   Microbiology at the TU Dresden for the screening of new antimicrobial
   substances and the next generation of antibiotics. The system enables
   biologists to screen agent combinations faster and use the gained image data
   to feed new deep-learning algorithms.

 * What, Where, How and Why? - Case Studies on Implementation of Lab of the
   Future Technologies in Discovery Life Sciences
   Umesh Katpally, Novartis
   
   There have been a good number of articles written, presentations made and
   webinars broadcasted on Lab of the Future (LoTF), including topics such as
   digitalization and automation, for the past few years. We continue to read,
   listen and discuss such topics even today and will continue to do so because
   the Future is always about looking ahead. In this presentation, we will look
   into the past few years to understand where we are currently in the context
   of implementing LoTF ideas and how they have been implemented. We will also
   look at what are some of the LoTF ideas that have not come to fruition and
   why. Based on this can we anticipate what new LoTF ideas will become into
   being in the years ahead?

DATA STRATEGY: SHARING AND REPURPOSING DATA

Session Chair: Marti Head, Ph.D., Oak Ridge National Laboratory (USA)

 * Adventures in Data Sharing
   Marti Head, ORNL
   
   To democratize access to integrated data and transform our ability to make
   data-driven decisions, organizations must break down barriers between siloed
   data producers and foster a culture of open-hearted sharing of richly
   contextualized data. This talk will unearth adventures in
   cross-organizational data sharing, with storytelling and lessons learned over
   my years at GlaxoSmithKline Pharmaceuticals and Oak Ridge National
   Laboratory.

 * To Blockchain or not to Blockchain? Practical Applications, Benefits and
   Considerations of Blockchain Technology in Laboratory Workflows
   Patrick Cullen, Yahara Software
   
   In today’s world, no lab is an island. Samples, protocols, data, and ideas
   need to be shared seamlessly within organizations and across scientific
   communities to streamline the path to impactful progress. Also, the vast
   amounts of data generated for each sample can be overwhelming to analyze and
   maintain securely. Lastly, determining if data and results can be compared
   apples-to-apples between different protocols, studies, or organizations can
   be almost impossible.
   
   In this talk, we will provide an overview of how blockchain can be a tool to
   achieve immense improvements in sample accessioning, data transfer, and
   workflow compliance using the ledgers inherent in the technology. Application
   of blockchain to these workflows can provide advantages such as elimination
   of ID duplication, reduction of missing or incorrect data, and decreased
   workflow errors and omissions, among others. When different organizations are
   part of the same blockchain, data integrity, protocol choice, and protocol
   adherence are transparent to all organizations involved. This shared
   technological ecosystem decreases barriers to communal data, study, and
   protocol repositories and increases the likelihood of productive
   collaboration.
   
   Despite these benefits, the implementation of blockchain technologies is not
   without challenges, requiring new tools, strategic thinking, and updated
   approaches. To assist participants in evaluating opportunities for employing
   blockchain technologies within their own laboratories, we will provide an
   overview of some considerations organizations should take into account, offer
   practical steps to evaluate potential blockchain use cases, and discuss
   potential limitations of the technology.

 * Enabling "Bench-to-Bedside" with FAIR data
   Viral Vyas, Bristol Myers Squibb
   
   Translational Medicine (“bench-to-bedside”) is a multidisciplinary field
   focused on developing new therapies and procedures that extend and enhance
   human life. Collaboration with internal and external labs is at the very core
   of this effort which poses numerous challenges in fulfilling its promise.
   Biomarker data from thousands of patients and multiple indications need to be
   collected, collated with clinical data, analyzed and visualized. Data needed
   to gain insights is often hard to find, incomplete, opaque, lacks conformance
   and governance. Bristol-Myers Squibb has embarked on a set of bold strategic
   initiatives dubbed “Digital Health - Sage” to tackle these challenges head on
   by using FAIR (Findable, Accessible, Interoperable, Reusable) data principles
   as a guide. Five key capabilities were delivered as part of the Digital
   Health – Sage effort: data lake, data catalog, analytics environment, search
   and visualization, data access and governance. The presentation will detail
   business challenges, technology solutions, and lessons learned.

 * Machine Learning is the Easy Part
   Lauren DeMeuse, Roam Analytics
   
   Data silos are prevalent across healthcare organizations, with challenges
   from people, process and technical capabilities. Once that data is shared and
   in a usable format, there's many new machine learning capabilities that can
   unlock improved data-driven decision making and shared context. This
   presentation will cover some of the typical challenges to data sharing,
   strategies for overcoming, and opportunities for advanced analytics once
   available.

FROM INSTRUMENT TO DECISION: IMPROVED DECISION-MAKING ON COMPLEX DATA AT SCALE

 * Session Chair: Nicola Richmond, Ph.D., GlaxoSmithKline (UK)

 * Deep characterization of drug libraries by image-based profiling and machine
   learning
   Michael Boutros, German Cancer Research Center
   
   Images of drug-perturbed cells harbour a breadth of information about drug
   effects that can be extracted and interpreted by machine learning
   methodologies. When combined with genetic perturbations, such assays provide
   a powerful approach to identify agonists and antagonists of potential targets
   for therapeutic interventions. Chemical-genetic interactions thereby allow an
   in-depth characterization of small molecules and their context-dependent
   effects.
   
   We have established a screening platform using high-throughput pipelines for
   experimental and analysis workflows which allows us to screen libraries with
   thousands small molecules. Using a simple staining procedure, termed
   cellmorph, we extract multiparametric profiles describing overall changes in
   cellular morphology and cell behavior using automated image analysis to
   enable a deep phenotypic profiling of small molecules and other
   perturbations.
   
   Here, we phenotypically measured chemical-genetic interactions between
   several mutant cell lines carrying single-gene knock outs and several
   thousand small molecules. Unsupervised clustering and statistical modeling of
   chemical-genetic interactions revealed promising interactions including
   synthetic lethality and resistance. We could further show how the phenotypes
   of mutant cell lines and small molecules can be quantified with machine
   learning classifiers, allowing a direct scoring and interpretation of
   drug-induced phenotypes. Importantly, the trained classifiers also
   efficiently quantified dosage-dependent effects of drugs. Furthermore, we
   will show how to apply image-based phenotyping to predict drug resistance and
   sensitivity in patient derived organoids.

 * Image-Based Cell Phenotyping Using Deep Learning
   Samuel Berryman, University of British Columbia
   
   The ability to phenotype cells is fundamentally important in biological
   research and medicine. Current methods of phenotyping cells rely primarily on
   flow cytometry to detect specific fluorescent markers. There are many
   situations where this approach is undesirable, such as problems with
   availability, specificity, cross-reactivity and cost of phenotyping markers.
   Furthermore, the number of markers required can increase the complexity, may
   exceed the detection limit, and even activate or decrease the viability of
   some cells. Finally, cells that are non-spherical or are too few are
   sometimes not compatible with flow cytometry. For these reasons, alternative
   methods for phenotyping are sought after, with the focus on live-cell
   imaging. Here, we investigate the potential to develop an “electronic eye” to
   phenotype cells directly from brightfield and non-specific fluorescence
   microscopy images.
   
   Cells from ten cancer cell lines (MCF7, MDA-MB-231, LNCaP, PC3, U2OS, HCT,
   THP-1, HL60, Jurkat and Raji) were non-specifically stained to identify their
   nucleus (Hoechst), cytoplasm (Calcein green), and actin filaments (SiR
   actin). Cells were dispensed into 96-well glass-bottomed imaging plates and
   then imaged at 10X using brightfield and three fluorescence channels. The
   microscopy images were segmented into four-channel 51x51 pixel images each
   containing a single cell. The segmentation process used the DAPI channel for
   locating individual nuclei and then used a combination of the other channels
   to ensure other cells, or debris were not in close proximity.
   
   To phenotype the cells, we developed a convolutional neural network (CNN)
   consisting of a four-channel input, four convolutional layers, one
   max-pooling layer, five fully connected layers, two dropout layers and seven
   batch normalization layers. ReLU was used as the activation function
   following each convolution or fully connected layer. Each node in the final
   fully-connected layer represented the probability of the imaged cell
   belonging to one of the known cell-lines. Softmax was utilized as a
   cross-entropy classifying error function for back-propagation during
   training.
   
   Our CNN was trained over 20 epochs with Adam optimization and dropout to
   avoid overfitting and rotational data augmentation to expand the dataset.
   Using five-fold cross-validation, we show that the CNN was able to recognize
   each cell line with a 94% average accuracy. Our results demonstrate the
   ability to use deep-learning to phenotype cells directly from microscopy
   images without specific markers. This capability will be valuable for
   situations where phenotyping markers are unavailable or the cell sample
   cannot be stained (such as before therapeutic use). We envision this approach
   to be a general method for identifying cell types directly from image data to
   identify the emergence of phenotypic shifts or new cell types.

 * Assessing Biological Diversity of a Compound Collection Using High-Throughput
   Cellular Imaging and No Ground Truth
   Yusuf Roohani, GlaxoSmithKline
   
   Image-based profiling of cellular phenotypes has emerged as a powerful source
   of information for comparing chemical and genetic treatments. This has opened
   the door to interrogating the biological impact of a chemical collection at a
   scale that was not possible before. However, optimizing models to interpret
   these images generally requires ground truth labels for the mechanism of
   action that are difficult to generate at scale using conventional techniques.
   Most often what is available for compounds in the discovery stage is the
   nominal target but that does not capture primary and off-target effects.
   Moreover, plate, batch and instrument variation can complicate the transfer
   of methods and analyses across different datasets or instruments limiting the
   utility of public data for this purpose. Thus, research groups are compelled
   to build data analysis methods using the same instruments and protocols that
   they would apply to their own data, even in the absence of corresponding
   ground truth. In this talk, we describe a reliable system for discerning and
   labeling distinct image-based cellular phenotypes in such a scenario. We
   cover several methods for feature extraction (hand-engineered features, deep
   learning) and analysis (clustering, similarity metrics, hit calling,
   correcting batch effects). Most of our methods are based on the central
   principle that biological replicates exhibit identical phenotypes. We run a
   follow-up assays to validate our results.

 * Interpreting AI Models Trained on High-Content Microscopy Data
   Oren Kraus, Phenomic AI
   
   The increasing popularity of high-content screening (HCS) and phenotypic
   profiling in preclinical drug discovery is generating enormous amounts of
   complex imaging data. The flexibility of these imaging-based assays allows
   researchers to quantify many different biological processes using a single
   technology. Examples include examining nuclear translocation of proteins,
   internalization of receptors, and morphological changes in response to tens
   of thousands to hundreds of thousands of treatments. Despite the experimental
   throughput of HCS, analyzing and interpreting HCS imaging data remains a key
   bottleneck in utilizing these systems. Scientists often need to collaborate
   closely with computer vision experts and data scientists to extract
   informative measurements (i.e. features) from imaging data and design
   customized analysis pipelines for each new assay. Machine learning provides a
   unique opportunity to automate and accelerate many of the steps involved in
   analyzing HCS screens.
   
   Recent results have shown that deep learning, specifically deep convolutional
   networks (CNNs) trained directly on raw pixel data, outperform existing
   approaches at classifying and clustering cellular phenotypes. The tradeoff
   often associated with these methods is the lack of interpretability of
   predictions made by deep learning models. We’ve designed several novel
   machine learning process for HCS that prioritize interpretability, by
   highlighting regions in the image that are responsible for the model’s
   predictions. These models combine fully convolutional neural networks,
   typically used for image segmentation, with convolutional multiple instance
   learning (convMIL) to aggregate predictions spatially across fields of view.
   Additionally, we’ve correlated predictions made by convMIL models with
   features extracted from individual cells using traditional feature extraction
   based analyses. Combining these two methods provides an additional layer of
   model interability by automatically indicating which features are changing
   most significantly between classes predicted by the CNN. Finally, we’ve
   developed a novel approach for exploring single-cell phenotypes in HCS
   screens using weakly-supervised learning models combined with an interactive
   tool for exploring phenotypes. Weakly supervised models are CNNs trained to
   predict every unique condition in an HCS screen based on image crops of
   single cells. Once the model is trained, a feature vector is extracted for
   every cell in the screen based on outputs from intermediate layers in the
   CNN. These feature vectors are then converted to 2D using dimensionality
   reduction techniques like t-SNE and UMAP. The interactive scatterplot we’ve
   built allows scientists to explore this 2D space while being able to see what
   individual cell phenotypes look like and which treatment conditions are
   common in different clusters that appear. We’ve used this tool to discover
   antibodies and compounds that are active in multiple assays that can include
   multiple cell-types and 3D culture systems. Taken together, these approaches
   significantly accelerate and improve phenotypic discovery programs.

Back to Top


DRUG TARGET STRATEGIES



This track is generously sponsored by MSD. 

Track Chairs: Margaret Scott, Ph.D., Genentech (USA) and Tim Wigle, Ph.D., Ribon
Therapeutics (USA) 

PROTEIN HOMEOSTASIS: NEW DRUG DISCOVERY DIRECTIONS

Session Chair: Robert Blake, Ph.D., Genentech (USA)

 * Leveraging Intrinsic Target Degradation
   Delphine Collin, Cedilla Therapeutics
   
   Protein degradation is a very effective way to inhibit target activity and
   prevent its scaffolding function. At Cedilla, we are focusing on small
   molecules that directly or indirectly regulate the homeostasis of a protein
   of interest. This presentation will cover some of our strategic approaches to
   direct and indirect degradation and the contribution of biomolecular sciences
   (biophysics, biochemical, structural biology, molecular dynamics…) to this
   novel path in drug discovery.

 * A Homogeneous Cell-Based Membrane Potential Assay to Identify Compounds That
   Promote Readthrough of Premature Termination Codons in the Cystic Fibrosis
   Transmembrane Conductance Regulator Ion Channel
   Emery Smith, Scripps Research Institute Florida
   
   Cystic fibrosis (CF), an inherited genetic disease, is caused by mutation of
   the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) gene, which
   encodes an ion channel involved in hydration maintenance via anion
   homeostasis. Nearly 5% of CF patients possess one or more copies of the
   G542X, which results in a stop codon at residue 542, preventing full-length
   CFTR protein synthesis. Identifying small molecule modulators of mutant CFTR
   biosynthesis that affect “readthrough” of this stop codon, or premature
   termination codon (PTC) to synthesize a fully functional CFTR protein
   represents a novel target area of drug discovery. We describe the
   implementation and integration for large scale screening of a homogeneous,
   miniaturized 1536-well functional G542X-CFTR readthrough assay. The assay
   utilizes HEK293 cells engineered to over-express the G542X-CFTR mutant, whose
   functional activity is monitored with a membrane potential dye. Cells are
   co-incubated with a CFTR amplifier and CFTR corrector to maximize mRNA levels
   and trafficking of CFTR, such that compounds that allow translational
   readthrough and synthesis of functional CFTR chloride channels will be
   reflected by changes in membrane potential in response to cAMP stimulation
   with forskolin, and CFTR channel potentiation with genistein. Assay
   statistics were excellent with Z’ values of 0.69±0.06 despite a S:B of
   1.19±0.04. As further evidence of HTS suitability, we completed an automated
   screening of 666,120 compounds, identifying 7,761 initial hits. Following
   secondary and tertiary assays, we have identified 188 confirmed hit compounds
   with low and sub-micromolar potencies. Thus, the assay has integrated the
   advantages of a phenotypic screen with high throughput scalability to
   identify new small molecule G542X-CFTR readthrough modulators.

 * Monovalent Versus Bivalent Degraders
   Robert Blake, Genentech
   
   The emerging drug design strategy based on inducing target protein
   degradation offers the potential of drugging classes of proteins not
   previously thought to be druggable. Furthermore, the magnitude of effect for
   these agents is not limited by receptor occupancy and the duration of effects
   can persist beyond drug exposure. The current design of protein degraders is
   more commonly based on bivalent molecules, which consist of a ligand for the
   target protein linked to a ligand for a ubiquitin ligase (such as VHL, CRBN
   or XIAP). Due to their bivalent design, such drugs typically have a higher
   molecular weight than classic small molecule drugs and may present some
   non-ideal properties as drugs. An alternative monovalent degrader strategy is
   exemplified by the group of drugs termed SERDs (selective estrogen receptor
   degraders), for example, fulvestrant. The molecular structures of SERDs are
   typically designed around a receptor-ligand and a “degradation tail”, whose
   presence results in the degradation of the estrogen receptor. We have
   recently reported that this monovalent strategy can also be applied to the
   bromodomain and extra-terminal (BET) family, with the example of the
   monovalent BRD4 degrader GNE-0011. We will use examples of monovalent and
   bivalent degraders of BRD4 to compare these two complimentary degrader
   strategies.

 * Exploring protein homeostasis using DELPhe assays
   Kandaswamy (Swamy) Vijayan, Plexium
   
   Modulating protein homeostasis using small-molecules is an exciting new area
   for therapeutics discovery. Inducing target degradation using bifunctional
   molecules has been the predominant approach to this field. We will describe a
   set of new tools that allows us to probe E3 ligase interactions without
   requiring ligands to targets apriori. Phenotypic assays that use DNA-encoded
   libraries (DELPhe) can scan chemical space for compounds that modulate E3
   ligases in specific and interesting ways. Examples of both single-target and
   multiple-target perturbations identified on DELPhe will be presented,
   including a machine learning formalism to evolve pleiotropic perturbations
   towards desirable goals.

DRUGGING RNA

Session Chair: Margaret Porter Scott, Ph.D., Genentech (USA)

 * Opening New Frontiers of Biology with RNA-Targeted Small Molecules
   Jessica Friedman, Arrakis Therapeutics
   
   The identification of drug-like small-molecule medicines that directly bind
   to RNA and modulate the biological function of RNA will vastly increase our
   therapeutic target space. RNA folds into structures that have diverse pockets
   into which small molecules can selectively bind. At Arrakis Therapeutics, our
   main focus is to identify small molecules that selectively bind RNA
   structures in critical regulatory regions of mRNA to modulate the expression
   of otherwise undruggable proteins. Our platform enables the analysis of RNA
   folding in vitro and in vivo as well as the identification of structures and
   pockets within an RNA. Drug-like compounds that bind these pockets are
   identified through high-throughput screening, followed by diverse biophysical
   assays to confirm and characterize binding such as SEC-MS, SPR and NMR. The
   biological impact of these compounds is evaluated in cell-based assays. The
   discovery of compounds that act via RNA binding is a new approach that has
   the potential to address previously intractable molecular targets and
   diseases.

 * Targeting Structurally and Functionally Diverse RNAs with Druglike Small
   Molecules
   Jay Schneekloth, NIH
   
   Recent estimates indicate that greater than 85% of the human genome is
   transcribed into RNA, yet just 3% of these transcripts code for protein
   sequences. Coupled with an increased knowledge of the noncoding functions of
   RNA and improved technologies for RNA structure determination, this
   information has given rise to interest in RNA as a therapeutic target for
   small molecules. In this presentation, I will discuss my group's recent
   efforts to understand RNA-small molecule interactions. We have developed a
   high throughput Small Molecule Microarray (SMM) screening platform, which we
   use to rapidly screen and profile RNA-binding small molecules. Further, I
   will discuss in detail several targets we have studied. I will also discuss
   our efforts toward the structure-guided design of RNA-binding molecules and
   the potential for future development.

 * Translating RNA Sequence into Lead Small Molecule Medicines and Progress
   Towards Small Molecule Antisense
   Matthew Disney, Scripps Research 
   
   A major challenge in Medical Science has always been capturing targets for
   drug development. The state-of-the-art in targeting of RNA is the use of
   oligonucleotide-based modalities that target RNA sequence. Our focus over the
   past 15 years has been on developing technologies to decipher which cellular
   RNAs are “druggable” targets for small molecules and which small molecules
   can target them, serving as lead medicines. I will describe advances in the
   area of Small Molecules Interacting with RNA (SMIRNAs), including a
   sequence-based small molecule rational design tool dubbed Inforna. This
   approach allows sequence-based design principles for SMIRNAs of which only
   oligonucleotides have been previously designed from the sequence. Inforna has
   enabled the design of SMIRNAs against RNAs that cause hard to treat cancers
   and incurable genetically defined diseases that have no known treatment. We
   will describe these compounds and their implications for leveraging known
   biology to advance lead medicines and also their implications as chemical
   probes to understand previously unknown RNA biology.
   
   I will also describe the development of approaches that allow for targeted
   degradation of RNAs in cells and animals by using SMIRNAs. For example, we
   have developed an approach that allows small molecules to recruit cellular
   nucleases to an RNA target to cleave it selectively and
   sub-stoichiometrically. Collectively, these studies show that small molecules
   can be designed to target RNA by using sequence-based design to deliver
   efficacious compounds targeting RNA including targeting the RNA for enzymatic
   destruction in cells and animals.

 * Development of a Novel High-Throughput Screening Approach to Target Specific
   RNAs
   John Joslin, GNF
   
   There is a growing appreciation for the many roles that structured RNAs play
   in disease progression. Accordingly, significant efforts are being devoted to
   identify low molecular weight compounds that specifically target these RNA
   molecules. These efforts may expand the number of therapeutic targets for
   disease intervention. While several groups are developing antisense
   oligonucleotides or siRNAs to target RNA, we sought to identify low molecular
   weight compounds that have desired drug-like properties, oral
   bioavailability, while retaining target specificity and potency. To
   accomplish this goal we have devised novel biochemical assays that are
   amenable to ultra-high-throughput screening. As a proof of concept, we have
   used these assays to identify compounds that bind well-characterized
   structured RNA, such as aptamers and ribozymes. For example, we developed
   screens that target the theophylline aptamer, which binds theophylline with
   high affinity and specificity. Through these screens, we identified
   theophylline, as well as several related molecules that bind the theophylline
   aptamer with an affinity that matches those reported in the literature. We
   also identified several compounds that are distinct from the theophylline
   scaffold, some of which have affinities exceeding that of the cognate ligand.
   To date, we have screened over 3 million compounds in these assays and have
   begun to understand the power and limitations of running HTS campaigns that
   target RNA. This presentation will highlight the assay development, screening
   results, and a view of what is required to carry out high-throughput
   screening campaigns that purposively target RNA.

ADVANCES IN CELLULAR TARGET ENGAGEMENT 

Session Chair: Tim Wigle, Ph.D., Ribon Therapeutics (USA)

 * Re-Evaluating Kinase Inhibitor Selectivity and Residence Time in Living Cells
   with Energy Transfer
   Matthew Robers, Promega
   
   I will describe the application of an energy transfer technique (NanoBRET)
   that enables an approach to broadly profile compound fractional occupancy and
   residence time against a variety of target classes inside intact, living
   cells. Using this method, a broad-spectrum evaluation of compound engagement
   can be measured against over 300 full-length human kinases in live cells.
   Target engagement potencies correlate strongly with potencies using more
   traditional pathway analysis readouts, thus providing a platform to establish
   structure-activity relationships (SARs) for kinase chemical probes or lead
   drug molecules.
   
   In live cells, we have uncovered a surprising spectrum of intracellular
   activity for certain cyclin-dependent kinase inhibitors (CDKi’s) and PROTACs,
   offering opportunities for repurposing some chemotypes as selective chemical
   probes for understudied kinases. We further evaluate opportunities for
   achieving target selectivity under non-equilibrium cell culture conditions,
   via protracted target residence time or PROTAC-mediated degradation. Here, we
   describe a broadly applicable approach for evaluating existing and novel
   chemical matter for selectively engaging CDKs in living cells.

 * A Bespoke Screening Platform to Study Mono (ADP-ribosylation)
   Tim Wigle, Ribon Therapeutics
   
   Mono(ADP-ribosylation) (MARylation) and poly(ADP-ribosylation) (PARylation)
   are post-translational modifications deposited on multiple amino acids, and
   emerging evidence suggests they are also deposited onto nucleic acids. There
   are 12 mono(ADP-ribose) polymerase (monoPARP) enzymes and 4 poly(ADP-ribose)
   polymerase (polyPARP) enzymes that use nicotinamide adenine dinucleotide
   (NAD+) as the ADP-ribose donating substrate to generate these modifications.
   While there are approved drugs and clinical trials on-going for inhibitors of
   the enzymes that deposit PARylation (specifically PARP1 and PARP2
   inhibitors), MARylation is gaining recognition for its role in immune
   function, inflammation and cancer, however there is a lack of chemical probes
   to study the function of monoPARPs in cells and in vivo. An important first
   step to generating chemical probes for monoPARPs is to develop screening
   assays to enable determination of potency and selectivity of inhibitors
   during the hit finding and lead optimization phases. Complicating the
   development of enzyme assays is that the substrates for the majority of the
   monoPARPs are unknown, and even for the ones with identified substrates, it
   is uncertain how they engage their substrates. Here we describe the
   development of multiple family-wide approaches to developing robust
   high-throughput monoPARP assays that overcome this lack of knowledge around
   their substrates.

 * CETSA®-HT: Enabling a New Paradigm in Hit Discovery
   Kirsten Tschapalda, AstraZeneca
   
   High throughput screening (HTS) cascades have evolved to ensure that
   high-quality hits can be identified from large screening collections.
   Traditionally, most primary screens focus on the identification of modulators
   of catalytically active sites, while target engagement assays are placed
   further down the cascade. Well established technologies like
   competition-based assays, affinity selection technologies or differential
   scanning fluorimetry (DSF) depend on the availability of protein which is
   tested in a non-native biochemical setting. Therefore, one of the main
   concerns when initiating an HTS cascade remains the demonstration of target
   interaction within a relevant cellular environment. The use of cellular
   assays during primary screening and the HTS cascade presents an alternative.
   However, cell-based screens can easily become very complex, risk off-target
   effects and thus often require time-consuming target deconvolution of pathway
   hitters. To date, there has been no single technology that can demonstrate
   cellular target engagement in a suitable format for HTS primary screening.
   The cellular thermal shift assay (CETSA®) can act as an interface between
   this classic biochemical-cellular screening dichotomy. CETSA® facilitates
   label-free screening in disease-relevant cells while approaching the ease of
   biochemical assays. In an isothermal setup, full assay plates are heated to a
   set point within the target protein melting curve. While most proteins unfold
   and precipitate upon this heat-shock, a characteristic of protein-ligand
   interaction is induced thermal stability. The remaining stabilized protein
   can subsequently be detected with a pair of anti-species antibodies in an
   AlphaScreen® system. This high throughput (HT) CETSA® format allows large
   numbers of compounds to be tested in an HTS setting. Here, we report the
   development of two CETSA®-HT assays along with the application of this
   technology in HTS for the first time. This has been enabled following the
   recent agreement between Pelago Bioscience and PerkinElmer to streamline
   CETSA®-HT into validated kits and to offer support in the assay development.
   In the Global High Throughput Screening Centre of AstraZeneca, we are
   exploring the potential and the feasibility of CETSA®-HT for large scale HTS
   campaigns ( >0.5M compounds). These datasets indicate the future impact
   CETSA®-HT will have in hit identification. This is particularly timely given
   the expanding interest across drug discovery groups in new target protein
   classes. With new modalities like PROTACS (proteolysis targeting chimera)
   non-catalytically active proteins can now therapeutically be targeted.
   Utilizing CETSA®-HT to identify target engagement in cellular environments
   early during primary screening could shift the paradigm of hit finding.

 * Targeting Engagement by Utilising CETSA in Drug Target Studies
   Laurence Arnold, Pelago Bioscience
   
   Target engagement is a fundamental paradigm in drug discovery. A significant
   portion of projects fail to reach the clinic due to lack of efficacy or
   failure to show the lead candidate is interacting with the intended target in
   a more complex environment. Using the proven CEllular Thermal Shift Assay
   (CETSA) technology to measure target engagement in various matrixes, such as
   tissues, intact cells or lysates is increasingly common for SAR studies and
   lead optimization. It is also possible to inform early-stage programs with
   CETSA technologies and investigate in-situ ligandability. This study looks to
   investigate a well-established oncology target using CETSA high throughput
   platform (HT). CETSA HT measures direct target engagement through the
   interaction of the protein and molecule after a heat challenge, with
   versatile readouts applicable to HTS and miniaturization for robotic
   platforms. Assay development for CETSA HT includes the development of HTS
   off-the-shelf kit formats and in this project, investigations into target
   engagement of fragments within a cellular environment. These target
   engagement studies are not reliant on in-vitro and often abstract functional
   screens or methods, often limiting to certain protein classes.

Back to Top


MICRO- AND NANO TECHNOLOGIES

Track Chairs: Amar Basu, Ph.D., Bioelectronica Corporation (USA) and Elodie
Sollier, Ph.D., Benkei (France)

MICRODEVICES FOR HIGH-THROUGHPUT CELLULAR AND BIOMOLECULAR PROFILING

Session Chair: Alex Shalek, Ph.D., MIT (USA)

 * Enabling Precision Medicine: Pipetting at Single-Cell Resolution
   Georges Muller, SEED Biosciences SA
   
   Single-cell isolation is essential in stem cell biology, cancer research and
   biotechnology among others. For example, to ensure quality, safety and
   efficacy of the biotherapeutic product, companies shall demonstrate that each
   new recombinant cell line has been cloned from a single progenitor cell (WHO,
   2014). Because available methods for cell cloning do not provide fully
   traceable cells yet, companies may waste up to 50 weeks in clonal validation.
   To solve this issue, we have developed an automated impedance-based pipetting
   robot for single-cell dispensing, allowing for traceable cloning of single
   cells. This technology permits the efficient and gentle isolation of
   industrial cell lines as well as rare and fragile stem cells and cancer
   cells, at a single-cell resolution, so that cells can be individually
   expanded in culture, transplanted downstream or analyzed by omics assays. We
   will present the technology and illustrate its key features through various
   case studies.

 * An Adaptable Microfluidic Platform for Single-Cell Pathogen Identification
   and Antimicrobial Susceptibility Testing
   Pak Kin Wong, The Pennsylvania State University
   
   Bacterial infections, such as bloodstream infections (BSI),
   ventilator-associated infection (VAI), and urinary tract infections (UTI),
   are a common cause of patient morbidity and mortality. Rapid identification
   of the causative pathogens and their antimicrobial susceptibility profiles
   will improve the clinical workflow for clinical management, accelerate
   clinical decision-making, and improve patient outcomes. However, definitive
   clinical microbiological analysis of samples obtained from patients requires
   several days, hindering proper management of infection and driving the
   overuse and misuse of broad-spectrum antibiotics. Novel precision
   technologies for rapidly identifying the pathogens and their antibiotic
   resistance are highly sought-after.
   
   To address this clinical unmet need, we develop a nanotube assisted microwave
   electroporation (NAME) technique for intracellular detection of
   species-specific bacterial 16s rRNA in 30 minutes. NAME allows
   amplification-free pathogen identification at the single-cell level. Unlike
   typical sensing techniques that lyse the bacteria and dilute the
   intracellular content, NAME directly detects species-specific regions of the
   16S rRNA inside the cells. Due to the small volume of a bacterium, the target
   molecule in the cell has a high effective concentration, which creates a
   strong signal for single-cell detection without amplification. Intracellular
   detection of bacterial 16S rRNA in viable cells also facilitates subsequent
   antimicrobial susceptibility testing (AST). By incorporating an adaptable
   microfluidic design, we demonstrate a phenotypic AST system that rapidly
   determines the existence of bacteria, classifies major classes of bacteria,
   detects polymicrobial samples, and identifies antimicrobial susceptibility
   directly from clinical samples at the single-cell level. The adaptable
   microfluidic system can dramatically accelerate the workflow of the
   microbiological analysis. Pathogen classification, which is based on
   microfluidic separation and microscopic inspection, eliminate the slow
   culture step. This approach rules out negative samples classify bacteria
   according to size and shape in as few as 5 minutes and identifies samples
   with multiple pathogens for polymicrobial infection diagnosis. By monitoring
   the bacterial growth directly, AST results can be reported in as few as 30
   minutes or in a time scale similar to the doubling times of the bacteria.
   
   In this study, we report the integrated microfluidic system for rapid
   pathogen classification and AST. We demonstrate the NAME technique for
   identifying bacteria that commonly cause BSI, VAI, and UTI. In collaboration
   with our clinical and industrial partners, we are developing an integrated
   ID-AST platform for the rapid diagnosis of bacterial infections. We pilot a
   study of 25 clinical urine samples to demonstrate the clinical applicability
   of the microfluidic system. The platform demonstrated a sensitivity of 100%
   and specificity of 83.33% for pathogen classification and achieved 100%
   concordance for AST. Our results demonstrate the analytical and clinical
   feasibilities of the integrated ID-AST platform for rapid microbiological
   analysis.

 * Building Tissue to Understand How Tissues Build Themselves
   Zev Gartner, UCSF
   
   The capacity of cells to self-organize into tissues is critical to their
   normal developmental and their ability to self-repair. Thus, a better
   understanding of how tissues self-organize will improve our ability to
   synthesize tissues and organs in the lab, and suggest new strategies to slow
   the breakdown of tissue structure that contributes to the initiation and
   progression of the disease. We are working to understand the mechanisms used
   by cells to self-organize robustly in the breast and gut, and how these
   programs are susceptible to the perturbations that underlie diseases such as
   cancer. We are also developing new single cell analysis technologies to help
   decode the logic of paracrine signaling networks that support the
   self-organization of these tissues.

 * Translating Single-Cell Genomics to the Clinic
   Alex Shalek, Massachusetts Institute of Technology
   
   While several methods exist for sampling tissues in clinical contexts,
   without high-fidelity tools for comprehensively profiling them, we are both
   limited in our capacity to understand how constituent cells and their
   interactions impact prognosis, and to select and develop precision
   therapeutics. Recent years have witnessed transformative and intersecting
   advances in nanofabrication and molecular biology that now enable deep
   profiling of low-input samples. Collectively, these afford new and exciting
   opportunities to study cellular heterogeneity, starting from the level of the
   single cell, and may unlock the diagnostic, prognostic, and discovery
   potential of clinical isolates. Illustratively, I will introduce how we can
   leverage single-cell genomic approaches – and, in particular, single-cell
   RNA-Seq – to explore the extensive functional diversity between cells,
   uncovering, from the “bottom-up,” distinct cell states and their molecular
   drivers. Moreover, I will discuss high-throughput experimental strategies and
   demonstrate how they can be leveraged to achieve the statistical power
   necessary to reconstruct intracellular circuits, enumerate and redefine cell
   states and types, and transform our understanding of cellular decision-making
   in health and disease on a genomic scale.

NOVEL SAMPLING STRATEGIES AND HIGHLY MINIATURIZED SYSTEMS FOR MASS SPECTROMETRIC
ANALYSIS

Session Chair: Daniel Austin, Ph.D., Brigham Young (USA)

 * Microscale Linear Ion Trap for Portable Mass Spectrometry
   Daniel Austin, Brigham Young University
   
   We present initial results from a high-aspect-ratio linear ion trap employing
   20-micrometer-wide electrodes patterned onto ceramic substrates, with a
   characteristic trapping dimension of 800 micrometers. In previous efforts, we
   showed that a variety of ion trap geometries can be made using assemblies of
   two ceramic plates, the facing surfaces of which are patterned with
   appropriately shaped electrodes. The present report shows the significant
   miniaturization of this approach. Mass spectra of organic compounds with this
   device have a resolution of 2-3 amu. These highly miniaturized analyzers are
   now being developed for portable GC-MS instrumentation. Aluminum electrodes
   were deposited onto one side of each ceramic substrate. Electrodes are
   wire-bonded to a printed circuit board, which connects with a capacitive
   voltage divider. Two plate-PCB assemblies are mounted in a sandwich
   configuration, with the trapping fields being established in the space
   between the plates. Prior to patterning, a tapered ejection slit, 166
   micrometers wide, was laser-cut into each substrate for ion ejection. The
   taper is critical to prevent ions from striking the inner wall of the slit
   and building up space-charge while allowing the thickness of the substrate to
   remain sufficiently thick for strength. Dipole resonant ejection of ions, in
   which the applied ejection waveform is phase-locked with the drive RF, was
   demonstrated by the use of special phase-tracking circuit. The alignment of
   the substrates was demonstrated using a set of 4 micropositioners (three
   linear and three angular). Low-power performance—essential for portable and
   hand-held mass spectrometers—was also demonstrated, with a maximum RF
   amplitude of 400 V at the highest point in the scan. The typical mass
   resolution of small organic compounds (toluene, xylenes) is 1.5 Da.
   Experiments using high molecular weight compounds (octofluorotoluene and
   perfluorotributylamine) showed typical mass resolution of 2-3 Da. The effects
   of higher operating pressure on mass spectra were also examined. Resolution
   decreased at pressures above 5 mTorr, but suitable spectra could still be
   obtained at pressures of up to 42 mTorr. Resolving power is decreased
   compared with the larger scale version of this device, possibly due to
   increased space charge. However, the signal to noise ratio is largely due to
   the high aspect ratio of these traps—the ratio of the length to the
   characteristic trapping dimension is greater than 40, providing a large
   trapping volume.

 * A Benchtop Biochemical Analyzer: Microchip Capillary Electrophoresis Coupled
   to High-Pressure Mass Spectrometry (HPMS)
   J. Michael Ramsey, University of North Carolina, Chapel Hill
   
   Biochemical analysis needs are frequently addressed using liquid
   chromatography coupled to mass spectrometry (LCMS) in a centralized
   laboratory setting. While these systems can be quite versatile to address a
   broad range of biochemical measurement problems, they are correspondingly
   complex and require a trained operator to produce results. LCMS
   instrumentation also typically occupies a large footprint and requires
   utilities beyond a simple power outlet. Our laboratory has been pursuing
   miniaturized versions of liquid phase separation systems and mass
   spectrometers for over two decades. We are combining these two technologies
   to demonstrate a compact benchtop analyzer that can address measurement needs
   in areas such as cellular biology, clinical diagnostics, and
   biopharmaceutical research and development that would normally be
   accomplished using LCMS.
   
   We have developed microfabricated capillary electrophoresis (microchip CE)
   devices with monolithically integrated nano-electrospray ionization (ESI)
   emitters that exceed the performance of conventional CE-ESI implementations.
   CE separations require ionic analytes, whereas LC can potentially separate
   either charged or neutral compounds. Biochemical species of interest are
   predominately ionic and CE systems outperform LC systems for separative
   performance, while the former can also be implemented more compactly with
   simpler components, e.g., voltage sources versus high-pressure pumps.
   Microchip CE has been used to separate ions as small as elemental species to
   intact monoclonal antibodies. One million theoretical plates of separation
   can be generated in one to a few minutes. Moreover, the microchip CE
   cartridge is easy to use and does not require any plumbing to connect the ESI
   emitter.
   
   We have also been involved in the development of a new form of mass
   spectrometry, HPMS, that can be implemented in a compact form as it operates
   at pressures several orders of magnitude higher than conventional MS, i.e.
   approximately 1 Torr. Operating at such pressures allows significant
   simplification of the vacuum system and the use of a vacuum pump that can
   rest in the palm of your hand. The mass analyzer in HPMS is a form of ion
   trap with sub-mm scale critical dimensions. We have theoretically and
   experimentally demonstrated that HPMS resolution can be increased by
   decreasing critical dimensions and correspondingly increasing the RF drive
   frequency.
   
   In this presentation, we will describe microchip CE and HPMS and the coupling
   of the two technologies to create a compact and useful biochemical analysis
   tool. The instrument implemented with a 96 well plate autosampler is
   approximately the size of a tower computer. Example applications such as
   monitoring bioreactor broth constituents will be presented.

 * A Small Footprint Ambient Ionization Enabled High-Throughput Chemical
   Detection System
   Brian Musselman, IonSense, Inc.
   
   Rapid analysis of the products of chemical reactions produced in
   high-throughput experiments (HTE) are completed by thermal desorption of
   sub-microliter volume samples into an ionizing gas. The heated ionizing gas
   completes the vaporization of the sample typically present in
   dimethyl-sulfoxide in 1-3 seconds per sample with rapid mass detection.The
   utility for Direct Analysis in Real Time (DART) for ionization of chemicals
   in the presence of aprotic solvents such as DMSO, and DMF has been employed
   to enable detection of those chemicals from sub-microliter volumes of the
   sample thus eliminating the need for sample dilution before analysis by
   LC/MS. The sub-microliter samples have been prepared by using several sample
   disposition method including low volume automated pipettor station and a high
   capacity disposable pin-tool. Using these devices we have chemicals present
   in concentrations appropriate for high throughput experiments that have been
   deposited onto a wire mesh surface which is then positioned between the DART
   source exit and the mass detector entrance for analysis. The use of small
   volume samples reduces the potential for matrix effiects by limiting the
   abundance of chemicals present in the ionizing gas. A rapid sampling of the
   small size droplets present on the sample supporting wire mesh enables
   continuous screening of the samples. We document the performance of each
   sampling method at analysis 1 per second to demonstrate the potential for a
   full 386-well sample plate analysis in under 10 minutes. Automated data
   analysis of the continuous collection of spectra in the data file is
   demonstrated using a file parsing software to permit the archival of the
   results. An outline of the overall workflow and its utility for simplifying
   the analytical effort in support of nanochemistry will be discussed.

 * ADE-OPP-MS: ESI-Mass Spectrometry-Based Bioanalytical Platform with
   Ultra-High Throughput
   Hui Zhang, Pfizer Inc.
   
   Recently, a new bioanalytical platform based on the coupling of acoustic
   droplet ejection (ADE) and open pore probe (OPP) technologies to mass
   spectrometry with standard electrospray ionization (ESI) ion source was
   introduced. Extraordinary performance has been demonstrated with this new
   platform, including plate-reader sampling speed, label free detection with
   MS, simplicity of assay development, just to name a few. As the pioneers to
   develop and apply this technology, our group has demonstrated the instrument
   capability and shared several seminal proof-of-concept studies supporting
   different areas of drug discovery including functional HTS, Drug-Drug
   Interaction (DDI), and other ADME applications in previous SLAS conference.
   With the commercialization of this new platform coming along, the technology
   had been further enhanced especially around system integration, automation,
   and robustness. We will be happy to report back the recent technology
   advancements, as well as the recent studies of the different screens Pfizer
   team has been working on to support live projects.
   
   One lipid based HTS assay has been developed and successfully applied for
   hits triage and SAR support. Challenges of the lipid sampling and handling
   will be highlighted, and solutions and performance highlights will be
   provided. Both false positive and false negative hits identified from other
   HTS means were effectively teased out taking advantage of the highly
   selective of MS detection. With the integration of liquid handling
   workstation (Beckman I7) and other key peripherals to the ADE-OPP-MS
   instrument and implementation of automation solutions, ultra-high throughput
   screening with mass spec became a reality and can provide up to hundreds of
   thousands of compounds per day throughput. Besides pharmacology advancements,
   high throughput parallels medicinal chemistry applications enabled by this
   ADE-OPP-MS technology will be shared. Ultra-low sample requirements provide
   by ADE technology enabled readouts with as little as 1uL reaction, making
   100X cost savings for the reagent and enabled much bigger chemical libraries
   to be made. Chemical reactions can be thoroughly evaluated with OPP-high
   resolution MS (HRMS) at second/reaction speed, 60-100X faster than the
   current paradigm. Such large and high-quality dataset enables the generation
   of big data around chemical reactions to feed in machine learning/artificial
   intelligence builds. Finally, biomarker analysis is another direction that we
   are interested in as another big advantage of the ADE-OPP-MS platform is that
   it can handle dirty sample matrixes with minimum sample preparation. We will
   show some recent example such as detecting N-methyl nicotinamide (NMN) from
   human urine samples or plasma pharmacokinetics (PK) studies where
   high-quality data (and equivalent to those obtained by conventional LC/MS
   method) were acquired without no sample preparation or just one step
   dilution; while the analysis time was effectively cut from multiple hours to
   a few minutes.

TECHNOLOGY DEVELOPMENT FOR MICRO AND NANOFLUIDIC DEVICES

Session Chair: Élodie Sollier, Ph.D., Benkei (France)

 * Limitations and New Methods in the Characterization of Microfluidic Devices
   for Manufacturing QC
   Maximilian Pitzek, Stratec Consumables
   
   The range of applications for microfluidic devices is constantly expanding
   and so are the challenges for manufacturing. There is always exciting for the
   development of new manufacturing methods, but the importance of new
   analytical methods is often underestimated by contract manufacturers. Here we
   present our latest advances in the metrology and derived QC of injection
   molded microfluidic devices. Especially in the development of nm-sized
   fluidic channels, µm-sized fluidic channels with nm-precision, and devices
   with a combination of inorganic and organic coatings we came across
   challenges that required new analytical methods. The questions range from a
   simple "how deep is a microfluidic channel after bonding", to "what
   manufacturing process has the biggest impact on channel roughness - from
   Mastering to the finished device". To answer these questions, we had to find
   new ways to characterize the different processes along the manufacturing
   chain. The analytical methods that we developed opened up the door to new
   types of devices, or made it possible to mass manufacture new high complexity
   consumables.

 * A Portable Quality Control Lab in The Era of Food and Beverage Craftsmanship
   Maciej Grajewski, SG Papertronics B.V. and University of Groningen
   
   In the past decade, we have observed a tremendous growth of interest in food
   and beverage craftsmanship due to new trends for sustainability and
   eco-friendliness. This has resulted in numerous exciting initiatives in food
   and beverage production. However, these initiatives have limited resources
   and thus struggle to maintain consistently high-quality processing.
   Preventing losses, financial and otherwise, caused by unpredictable events
   such as microbiological contamination is often challenging.
   
   Traditionally, these problems are tackled by quality-control (QC) protocols
   embedded in the production process. However, the implementation of QC often
   requires trained personnel and professional analytical equipment, with
   associated costs well beyond the budgets of small entrepreneurs. Therefore,
   it is a great opportunity for the rapidly growing field of portable
   (bio)chemical analysis to step in and offer a viable solution for this
   market. However, tests are often developed for trained chemists who are
   capable of proper sample acquisition and data interpretation. We propose an
   alternative technology that can accommodate common colorimetric tests, in a
   portable format that can be used by non-chemists. Moreover, the technology
   provides shorter sample-to-answer times for samples collected and analyzed
   directly at the production site by the craftsman. To achieve this, we utilize
   a patented sample concentration technology that enables quantitative
   analytical tests with enhanced sensitivity, combined with a sample
   acquisition system and data interpretation software.
   
   The sample concentrator used in this work was developed by rapid prototyping
   with a stereolithographic 3D-printer. The concentrator cartridge consists of
   a sample acquisition module for volumetric sampling with subsequent liquid
   transfer to a porous particulate column packed into a 3D-printed cartridge. A
   series of branched air ducts embedded in the sample concentrator guide
   pressurized air from a simple gas supply to a selected region of porous
   membrane fixed into the bottom of the cartridge. Liquid samples are reacted
   with reagents in the 3D cartridge and then concentrated on the membrane
   through evaporation of solvent by the gas. The analysis is subsequently
   carried out by colorimetry. Integrated software analyzes the test results and
   compares it to results stored in our database so that every craftsman can use
   our system without the need for extensive chemical training. Additionally,
   our cartridges provide these tests with better protection against
   contamination, with improved user-friendliness, and the possibility of
   combining multiple materials for one test.
   
   The described technology has been applied for QC testing in different
   branches of the food and beverage industry. Our technology contributes to the
   market of portable analysis because it provides a tool that can be applied
   outside an analytical laboratory, but with comparable results. This means
   shorter times between sampling and result, and, importantly, provides
   substantially better options for the small food-and-beverage entrepreneur to
   realize improved QC.

 * Single-Cell Hybrid Microfluidics as a Selection and Sorting Tool in the
   Mammalian Gene-Editing Pipeline
   Kenza Samlali, Concordia University
   
   Developing new engineered clonal cell lines is essential for loss-of-function
   studies, investigating protein function and unraveling signaling cascades and
   metabolic pathways. The current mammalian cell engineering pipeline of DNA
   delivery, selection, screening or sorting, and single-cell clonal expansion
   remains challenging and heavily relies on multiple expensive automated
   systems like a flow cytometer, cell sorters and colony pickers to increase
   experimental success.
   
   Our group has shown the use of digital microfluidics for automating
   gene-editing procedures (Sinha et al.,2018), yet these devices cannot still
   select for successful edits or sort cells out into single clones.
   
   Hybrid microfluidics combine the digital and droplet microfluidic paradigms
   in one device (Ahmadi et al., 2019), with electrode lined channels that can
   introduce more control over droplets in channels. In this talk, I will
   present four key results: First, we developed a hybrid microfluidic method
   for deterministic single-cell encapsulation. I’ll introduce a single-cell
   trapping array that is capable to achieve near-perfect one-cell-per-droplet
   encapsulation. Second, I will present the droplet operations that can be
   performed on this device. This includes precise on-demand droplet operations
   including releasing, merging and keeping single-cell containing droplets.
   This allows for dynamic assays of mammalian single cells, based on screening
   parameters that go beyond the traditional fluorescence based screening or
   sorting methods. All these operations, including encapsulation, can be easily
   performed with a graphical user interface. Third, I will describe the
   efficiency of cell trapping, cell encapsulation, droplet release and droplet
   keeping under different flow rates. Fourth, I will describe results that
   validate our platform for use in the mammalian cell engineering pipeline,
   using a breast cancer cell line (MCF-7) and a lung carcinoma cell line
   (NCI-H1299) as a model system. Specifically, I will show results related to
   encapsulation of a heterozygous and edited cell population and instant
   selection of transfected single-cells with subsequent generation of a clonal,
   isogenic edited cell line that could be expanded off chip.
   
   This novel microfluidic device avoids the use of flow cytometry, cell sorters
   or limited dilution experiments to establish monoclonal engineered cell
   lines. Furthermore, the device can operate with low cell counts, which can
   potentially be used to handle sensitive cell lines along with enrichment of
   rare populations.

 * Microfluidic circuits with fluid walls to study cell migration
   Cyril Deroy, Oxford University
   
   A microfluidic technology based upon the use of “fluid walls” was recently
   introduced to address the lack of uptake of traditional microfluidic devices
   in biomedicine. Reasons cited for this lack of uptake include technical
   complexity, high failure rates due to gas-bubbles altering flows and
   affecting cells in micro-channels, the questionable bio-compatibility of
   materials like polydimethylsiloxane used to make devices, the inaccessibility
   of cells growing in them, and – probably the most important – biologists
   cannot use their familiar culture dishes and microscopes. The new technology
   reshapes fluid interfaces between two immiscible liquids (cell-growth medium
   and a bio-inert fluorocarbon, FC40) in a standard cell-culture dish, to form
   arrays of isolated liquid chambers. Each aqueous chamber is separated from
   its neighbors by transparent liquid walls of FC40. At the microscale, these
   fluid walls prove to be strong, pliant, and resilient. We now extend this
   fluid-shaping technology to create complicated circuits that can have almost
   any imaginable 2D shape, and demonstrate the power of the approach through a
   range of dynamic biological assays, in which cells migrate up concentration
   gradients established by diffusion. In one, primary mouse macrophages from
   the bone marrow are imaged as they migrate over polystyrene towards a
   chemo-attractant. More complex circuits in which cells are given a choice
   between different competing chemo-attractants allow the analysis of the
   decisions those cells make. In another, a steep gradient over less than 100
   µm is generated by diffusion between two laminar streams as they flow through
   a conduit (width 500 µm, height 50 µm); bacterial cells (Pseudomonas
   Aeruginosa) growing in the conduit then migrate over glass towards an
   antibiotic. Finally, circuits with fluid-walls are built within wells of
   96-well plates, to demonstrate that this technology can be seamlessly
   incorporated into standard high-throughput workflows based on microplates.
   All these circuits are built in minutes on virgin Petri dishes or standard
   plastics. The fluid walls allow direct access to any part of the circuit,
   they can even be reconfigured during the experiment, and cells can be
   recovered through them at any stage for analysis.

Back to Top


MOLECULAR LIBRARIES

Track Chairs: Andrew Alt, Ph.D., University of Michigan (USA) and Guy
Breitenbucher, Ph.D., University of California San Francisco (USA)

SMALL MOLECULE LIBRARIES

Session Chair: Guy Breitenbucher, Ph.D., University of California San Francisco
(USA)

 * Integrated Phenotypic Screening and Chemical Proteomics to Discover Druggable
   Immunomodulatory Pathways
   Ekaterina Vinogradova, Scripps Research 
   
   Modern human genetics has underscored the important role that the immune
   system plays in many human diseases ranging from autoimmunity to
   neurodegeneration to cancer. Nonetheless, chemical probes are still lacking
   for many immunologically relevant proteins and protein classes. We have
   applied an integrated phenotypic screening and chemical proteomic strategy to
   globally map the druggable proteome of human immune cells. Leveraging
   extensive past knowledge on the special capacity of cysteine-reactive
   small-molecule electrophiles to perturb immune system function, we have
   profiled this class of compounds against thousands of cysteines in primary
   human immune cells and will present compelling evidence that this strategy
   has already identified hit compounds for diverse classes of proteins with
   genetic links to human immune disorders.

 * Doubling Down: Betting on the success of HTS & DEL libraries in parallel
   David Lancia, FORMA Therapeutics
   
   High-throughput screening (HTS) libraries and DNA-encoded libraries (DELs)
   are two key repositories from which active hit compounds can be identified.
   FORMA has implemented a screening paradigm that enables HTS and DEL in
   parallel to enhance the likelihood of discovering hits for a particular
   target. Utilizing both methodologies in parallel allows us to thoroughly
   sample an expanded chemical space compared to either individually as well as
   evaluate potentially unexpected mechanisms of action (MOA) for each target.
   We will present our current success using the combined approach as well as a
   retrospective analysis of how expanded chemical space and novel MOAs could
   have further enhanced our past success delivering lead compounds.

 * Automated Chemistry: From an Idea to Reality
   Michael Kossenjans, AstraZeneca
   
   The discovery of bioactive small molecules is generally driven via iterative
   design-make-purify-test cycles. Automation is nowadays routinely used at the
   purify and test stage of these cycles but still very rarely at the design and
   make a stage. However, recent advances in areas such as
   microfluidics-assisted and batch chemical synthesis as well as AI systems
   that improve a design hypothesis through feedback analysis, are now providing
   a basis for the introduction of greater automation into all aspects of this
   process.
   
   Here, we describe recent progress we have made with the build of a fully
   automated synthesis platform comprising all aspects of the make-purify
   workflow. A variety of chemistry transformations have been established on the
   platform allowing not only the rapid synthesis of compound libraries but also
   complex molecules via multistep sequences. The value and impact of our
   platform is illustrated in the context of specific case studies from the
   early phase drug discovery.
   
   We also consider the longer-term goal of realizing the fully autonomous
   discovery of bioactive small molecules through the integration of our
   automated synthesis platform with automated design and testing.

 * Library Of Compounds with Really Annoying Pharmacology (LOCRAP)
   Jarrod Walsh, AstraZeneca
   
   False positive results have long been the bane of High Throughput Screening
   (HTS) campaigns. Depending on the compound library tested, the target itself
   and the assay system employed these have been estimated to account for
   enrichments of up to 95 % in hit outputs. Many sources of undesirable hits
   exist whether they be technology artifacts, redox cycling compounds,
   inhibitors of coupled enzyme systems or any other from a myriad of
   mechanisms. One strategy to cope has been the development of so called
   ‘nuisance compound’ sets. Over the past decade numerous pharmaceutical
   companies acting in isolation, including AstraZeneca, have generated their
   versions. Whilst the rationale, composition, source and application of these
   decks have varied between organizations the end goal has remained constant.
   Each group has aimed to either minimize the prevalence of undesirable actives
   by optimizing assay design or build bespoke triage cascades capable of
   identifying them. This presentation details the process by which the
   AstraZeneca set was established and shows the impact it’s had on drug
   discovery projects. We explore how it differs from those created by some of
   our peers and finally introduce a new cross industry and academia initiative
   called LOCRAP. This Library Of Compounds with Really Annoying Pharmacology
   (LOCRAP) is a collaboration between AstraZeneca, Eli Lilly, Novartis, Pfizer,
   Broad Institute, NCATS and lead academics in the area Jonathan Baell (Monash
   University) and Mike Walters (University of Minnesota). By pooling our
   collective knowledge, the intention is to design and deliver an industry
   standard collection that will be available to all through a third-party
   commercial partner. This new set will cover multiple classes of problematic
   compounds and contain thoroughly validated and well-annotated examples. We’ll
   discuss how this has been achieved and what classes are currently included.
   We are actively seeking contributors with ideas on compounds to include or to
   act as beta-testing supporters once the collection is available. We hope to
   make the tools and resources available to large pharmaceutical companies an
   option for any organization interested in drug discovery no matter how large
   or small. From this arises the aspiration to globally improve the quality of
   assays employed for hit identification and subsequently success rates for
   discovery campaigns.

DNA-ENCODED LIBRARIES

Session Chair: Martin Matzuk, M.D., Ph.D., Baylor College of Medicine (USA)

 * Exploring Reliable and Cost-Effective DNA-Encoded Library Approaches for
   Developing Active Compounds
   Raphael Franzini, University of Utah
   
   Tagging combinatorial libraries with DNA barcodes allows using a simple
   affinity selection protocol to rapidly identify protein binders. A primary
   challenge with such DNA-encoded libraries (DELs) is how to design them to
   provide the effective screening productivity needed for the routine discovery
   of developable hits. The predominantly pursued approach is to make platforms
   of very large libraries of chemically complex compounds. While many success
   stories have proven the validity of this paradigm, it is unclear whether such
   DELs compare favorably to competing technologies with regard to
   return-on-investment. Moreover, the often heterogeneous synthesis yields of
   very large libraries and undersampling of DNA-barcodes impedes effective hit
   triaging. The cost of producing large DEL platforms and identifying hits from
   screening data is completely prohibitive for laboratories with limited
   resources. We, therefore, explore alternative library designs to find active
   compounds at lower DEL synthesis and lead-development costs. We custom-design
   DELs for specific target classes, and early studies have demonstrated that
   such libraries provide hits rapidly and economically. For example, a small
   and chemically simple DEL targeting NAD+-binding sites provided potent and
   target-selective hits for ADP-ribosyltransferases. Possible strategies for
   advancing early screening hits from such chemically simple libraries will be
   discussed.

 * Drug Discovery with DNA-Encoded Chemical Libraries
   John Faver, Baylor College of Medicine
   
   DNA-Encoded chemical Libraries (DEL) enable efficient screening of billions
   of drug-like small molecules for binding affinity to protein targets. With
   DEL, products of combinatorial chemical synthesis can be screened as a single
   mixture, because each compound is covalently linked to a DNA segment with a
   known identifying sequence. Affinity selection experiments are conducted to
   isolate small molecules that bind to protein targets under specific
   conditions, and DNA sequencing is used to identify binders and quantify
   enrichment. We have developed a DEL synthesis and screening platform in the
   Center for Drug Discovery at Baylor College of Medicine which utilizes novel
   on-DNA chemistries to generate both general and target-specific libraries.
   These libraries have produced hit series with robust structure-activity
   relationships for multiple target families. We have also developed enhanced
   data analysis strategies that allow for quantitative comparisons of
   enrichment from multiple screens, providing information such as target
   selectivity, locations of binding sites, and relative binding affinities.
   These strategies led us to the swift discovery of potent and selective hit
   compounds for a variety of targets with little additional medicinal chemistry
   optimization.

 * Off-DNA DNA Encoded Library Affinity Screening
   Amber Hackler, Scripps Research 
   
   DNA-encoded library (DEL) technology is emerging as a key element of the
   small molecule discovery toolbox. DELs are highly diverse collections of
   combinatorially synthesized small molecules (~300-800 Da) attached to very
   large encoding DNA molecules. During affinity selection, the DNA tag can
   participate in the binding interaction, leading to false positive results and
   confounding the investigation of nucleic acid-binding targets (e.g.
   polymerases, transcription factors). An ideal affinity screen would only
   interrogate the library member for binding. Here, we use solid-phase DELs and
   microfluidic screening to separate each DEL member from its encoding tag and
   detect target binding using laser-induced fluorescence polarization (FP). DEL
   beads and an FP probe (dye-labeled ligand) are encapsulated in water-in-oil
   emulsion droplets containing the macromolecular target. Inside the droplet,
   the DEL member is photochemically cleaved from the DNA-encoded bead and
   laser-induced FP is measured. If the photocleaved library member competes
   with the probe for target binding, the probe emission is relatively
   depolarized, triggering electrokinetic droplet sorting and collection for
   follow up DNA sequencing. We prototyped this screening mode using the
   receptor tyrosine kinase (RTK) discoidin domain receptor 1 (DDR1), which is
   overexpressed in many cancers (e.g., leukemia, brain). A fluorescein-labeled
   DDR1 ligand (discovered in a previous affinity-based DEL screen) was used to
   build the droplet-scale competition binding assay. The FP signal difference
   between droplets containing either DDR1 or DDR1 and unlabeled competitor (20
   µM) resulted in a Z’ of 0.58. After confirming assay robustness, a 67,100
   member solid-phase DEL of drug-like small molecules was screened for ligands
   of DDR1 using the droplet-scale competition binding assay. Of the
   high-priority hit structures, several known RTK inhibitor pharmacophores were
   identified, including azaindole- and quinazoline-containing monomers. Off-DNA
   DEL affinity screening is amenable to screening in cell lysate and more
   complex affinity-based interrogations, such as interactome perturbation, in
   addition to providing an avenue to conduct mechanism-based screening using
   DEL.

 * DEL out of Water
   Philip Dawson, Scripps Research
   
   The structural diversity of DNA Encoded Libraries has been limited since the
   hydrophilic, unprotected nature of the DNA tag severely limits the repertoire
   of compatible chemical reactions. Rather than pursuing the optimization of
   individual synthetic organic reactions for water compatibility, we reasoned
   that a general strategy for transferring DNA-substrates into organic solvents
   could significantly expand the structural diversity explored by DEL.
   Reversible absorption of macromolecules to a solid support (RASS) has
   facilitated peptide and protein modification, enabling the use of anhydrous
   solvents and multistep synthetic procedures. This RASS strategy was adapted
   for DEL through a polystyrene based, quaternary ammonium resin. Adsorption of
   DNA headpiece substrates to this resin was found to facilitate transfer to
   organic solvents such as DMA, THF, and CH2Cl2. This RASS approach for DEL has
   enabled the development of Ni mediated carbon-carbon (C(sp2)-C(sp3)) and
   carbon-heteroatom (C-N, C-S, C-P) cross couplings with broad substrate scope
   and with excellent DNA compatibility. The immobilization of the DNA has also
   facilitated the use of electrochemical transformations. This expanded scope
   of reaction conditions compatible with DEL library generation has the promise
   to contribute to the generation of conformationally diverse scaffolds with
   drug-like properties.

CHEMISTRY AI

Session Chair: Gerard Rosse, Ph.D., Dart Neuroscience (USA)

 * De Novo Drug Design with Chemistry-Savvy Machine Intelligence
   Gisbert Schneider, ETH Zurich
   
   Chemical creativity in the design of synthetic chemical entities with
   druglike properties has been the domain of medicinal chemists. At the same
   time, constructive machine learning models have been shown to autonomously
   sample drug-like molecules from chemical space without the need for explicit
   design rules. A machine learning method that combines a rule-based approach
   with a machine learning model was trained on synthetic routes described in
   chemical patent literature. This unique combination enables a balance between
   ligand-similarity based generation of innovative compounds by scaffold
   hopping and forward-synthetic feasibility of the designs. Prospective results
   demonstrate the capability of this hybrid machine learning model to capture
   implicit chemical knowledge from chemical reaction data and suggest feasible
   syntheses of new chemical matter. We will present various applications of
   molecular de novo design with machine intelligence, and discuss the
   advantages and limitations of these design concepts.
   
   Button, A., Merk, D., Hiss, J. A., Schneider, G. Nat. Mach. Intell. 2019, 1,
   307–315.
   Schneider, G. Nat. Mach. Intell. 2019, 1, 128–130.
   Schneider, G., Clark, D. Angew. Chem. Int. Ed. 2019, 58, 10792–10803.
   Schneider, G. Nat. Rev. Drug Discov. 2018, 17, 97–113.

 * Discovering Multi-Target Pharmacology of Drugs and Drug Candidates by 3D
   Target Models and Machine Learning
   Ruben Abagyan, University of California, San Diego
   
   Small molecule therapeutics have an extensive, and only partially known,
   multi-target pharmacology that defines both their beneficial and adverse
   effects. We have characterized these networks for all cancer drugs.
   Furthermore, a team of researchers at Molsoft and UCSD developed a set of
   thousands of models in which 3D models are combined with the machine-learning
   layer to predict the activity of any chemical against protein targets
   included in this panel. The models can be used to discover targets of any
   known drug or drug candidate, search for compounds with specific multi-target
   profile, repurpose drugs for a new indication or disease, or identify
   potential liabilities. Applications of a multi-profile approach are
   presented.

 * Applying Artificial Intelligence and Machine Learning Techniques and Cross
   Platform Communication to Enable Informatics Driven Experimentation
   Carleen Klumpp-Thomas, NIH/NCATS
   
   Over the past decade, there has been a shift away from the traditional static
   method of performing high throughput screening (HTS) against large chemical
   libraries where experiments are designed in advance, executed and then the
   generated data is processed. Traditionally, this results in additional rounds
   of biological validation, testing and lead compound follow up for medicinal
   chemistry. More recently there has been a movement to focus instead on an
   increased number of targeted chemical libraries and smaller initial HTS
   experiments which can be run more dynamically with the resultant data
   processed automatically and in near real time to initiate new biological
   experimentation and even automated chemical synthesis on the fly. To make
   this possible it is necessary to have an underlying software and messaging
   infrastructure that can connect informatics platforms that utilize Artificial
   Intelligence and Machine Learning techniques to design experiments that can
   then be transferred to physical systems to initiate new experiments or small
   molecule synthesis. NCATS has developed such a platform with the initial
   validation being used to perform dynamic assay optimization but which is
   extensible to far more complex experimentation types to move beyond
   automation and instead towards autonomy.

 * Case Studies in AI-Driven Drug Design
   John Griffin, Numerate, Inc.
   
   Breakthroughs in machine learning theory and practice, coupled with ready
   access to cloud based supercomputing resources and ever-increasing amounts of
   experimental data, are enabling truly AI centric processes for small molecule
   drug design wherein predictive models successfully substitute for laboratory
   assays throughout the Discovery critical path. This presentation will
   describe how diverse applications of machine learning techniques, ranging
   from multidimensional and multitask boosting to deep neural networks, can
   extract accurate, scaffold independent, ligand based predictive models for
   important phenomena: target binding, functional activity, selectivity,
   PK/ADME properties, and toxicity. Applications of these models will be
   illustrated with examples from therapeutic programs and discussed in terms of
   their potential to enhance success/reduce attrition in drug discovery.

Back to Top


PRECISION MEDICINE TECHNOLOGIES

Track Chairs: Kristen Brennand, Ph.D., Mount Sinai School of Medicine (USA) and
John Joslin, Ph.D., Genomics Institute of the Novartis Research Foundation (GNF)
(USA)

TOOLS TO RESOLVE DISEASE COMPLEXITY

Session Chair: John Joslin, The Genomics Institute of the Novartis Research
Foundation (USA)

 * Toward Precision Medicines for Rare Diseases
   Anna Greka, BWH/Harvard/Broad
   
   Intracellular accumulation of misfolded proteins causes toxic
   proteinopathies, diseases without targeted therapies. Mucin 1 kidney disease
   (MKD) results from a frameshift mutation in the MUC1 gene (MUC1-fs). Here, we
   show that MKD is a toxic proteinopathy.
   
   Intracellular MUC1-fs accumulation activated the ATF6 unfolded protein
   response (UPR) branch. We identified BRD4780, a small molecule that clears
   MUC1-fs from patient cells, from kidneys of knockin mice and from patient
   kidney organoids.
   
   MUC1-fs is trapped in TMED9 cargo receptor-containing vesicles of the early
   secretory pathway. BRD4780 binds TMED9, releases MUC1-fs, and reroutes it for
   lysosomal degradation, an effect phenocopied by TMED9 deletion. Our findings
   reveal BRD4780 as a promising lead for the treatment of MKD and other toxic
   proteinopathies. Generally, we elucidate a novel mechanism for the entrapment
   of misfolded proteins by cargo receptors and a strategy for their release and
   anterograde trafficking to the lysosome.

 * A Hydrogel-Enabled 3D Liver Fibrosis Model for High-Throughput Phenotypic
   Screening Applications
   Zhixiang Tong, GNF/NIBR
   
   Hepatic stellate cell (HSC) is one of the major cell types responsible for
   the progression of liver fibrosis. Inhibition of HSC activation (i.e.
   transdifferentiation from an inactive/quiescent state to a myofibroblast-like
   state) represents a compelling strategy for resolving liver fibrosis.
   Conventional 2D HSC culture cannot recapitulate the in-vivo 3D cell-ECM
   interactions and often causes undesirable cell activation due to the
   intrinsic mechanical properties of the plastic substrates. Hydrogel has
   emerged as a powerful tool for modeling the 3D cellular microenvironment in
   vitro, largely due to its chemical and physical versatility. To this end, GNF
   team has developed a comprehensive toolbox comprised of a variety of
   polyethylene, hyaluronic acid and gelatin-based hydrogel derivatives.
   Specifically for modeling HSC activation, we’ve identified a gelatin
   methacrylate (GelMA)-based hydrogel formulation that preserves the quiescent
   state of HSCs much better than 2D culture on tissue culture polystyrene, and
   precisely controls the pro-fibrotic signal (i.e. TGFβ) induced activation in
   3D. Such control offers optimal TGFβ induced response window superior to that
   of standard 2D culture, leading to robust assay performance in a highly
   miniaturized format. By leveraging GNF’s proprietary engineering/automation
   technologies, the 3D culture can be efficiently implemented in a 1536-well
   plate format with minimal inter-/intra-plate variations. Using a tool GNF
   compound set (with ~1800 compounds), our pilot high-content imaging (HCI)
   based phenotypic screen reveals an assay robust Z’ factor over 0.45 and a
   hit-picking rate ~3%. Overall, our automation-friendly, hydrogel-enabled 3D
   HSC assay can be readily scaled up either for primary screen or lead
   profiling applications, and the novel screening platform shown herein could
   offer broad utility to identifying novel targets or therapeutic modalities
   for diseases beyond liver fibrosis.

 * Development of Colorectal Cancer Patient-Derived Organoid-Fibroblast Models
   Suitable for Drug Testing
   Eliza Fong, National University of Singapore
   
   The advent of patient-derived organoid (PDO) technologies has greatly
   expanded the toolbox for drug discovery and personalized drug screening for
   several cancer types. While PDO models more closely represent the molecular
   characteristics and heterogeneity of patient tumors than traditional
   immortalized cancer cell lines, they are inherently limited in their ability
   to reflect the tumor microenvironment in vitro as they comprise exclusively
   of epithelial cells. The lack of stromal cells in PDO models, such as
   cancer-associated fibroblasts (CAFs), poses a major problem as these tumor
   microenvironmental components contribute to the various hallmarks of cancer
   and response to therapy. Particularly for colorectal cancer, CAFs comprise
   the majority of the tumor microenvironment and play important roles in cancer
   development and progression, from the regulation of cancer cell proliferation
   and stem cell maintenance to drug resistance. In this study, we addressed
   this problem by establishing in vitro conditions that robustly enable the
   co-culture of CRC PDO with patient-derived CAFs for controlled mechanistic
   studies and drug testing. We report the development of an engineered tumor
   microenvironment consisting of CRC PDO encapsulated within a well-defined
   three-dimensional (3D) hyaluronan-gelatin hydrogel and co-cultured with
   patient-derived CAFs. Basement membrane extracts (e.g. Matrigel)
   conventionally used for PDO culture exhibit batch-to-batch variability.
   Considering that the CRC extracellular matrix is high in hyaluronan and
   collagen I and that hyaluronan-based matrices are conducive for the culture
   of various human cancers, we hypothesized that hyaluronan-gelatin hydrogels
   may serve as a suitable alternative 3D matrix to support the culture of CRC
   PDO and CAFs. Through RNA- and whole-exome sequencing, we first show that
   these hydrogels are capable of maintaining the molecular characteristics of
   the original patient tumors in the cultured CRC PDO. Further, based on our
   findings that standard PDO culture medium poorly supports CAF viability, we
   developed a new co-culture strategy that robustly maintains the viability of
   both CRC PDO and CAFs for at least a week in culture. We found that in the
   absence of any growth supplements added to the co-culture, CAFs were able to
   maintain the growth of the cultured CRC PDO in the hydrogels. Lastly, we
   demonstrate that these CRC PDO-CAFs models are suitable for evaluating
   standard-of-care drugs, making them potentially very useful for realizing
   personalized cancer medicine.

 * Liquid Biopsy as a Tool in Ant-Cancer Drug Screening for Personalized Therapy
   Decision and Drug Discovery
   Kamran Honarnejad, Fraunhofer ITEM
   
   The use of circulating tumor cells (CTCs) isolated from liquid biopsy are
   already used to predict disease progression and survival in metastatic
   patients. However, the lack of robust drug screening assays has hampered
   their application in monitoring patient drug response/resistance and
   personalized therapy decision.
   
   We have developed a workflow to isolate tumor cells from pleural effusion and
   malignant ascites samples from metastatic lung and breast cancer patients and
   subjected them to medium scale drug screens against approved anticancer drug
   libraries. This approach allows the realization of personalized treatment
   decisions within less than a week by evaluating drug responses directly in
   patient-derived tumor cells obtained from liquid biopsy.
   
   In patients with no pleural effusion or malignant ascites, we have
   established another workflow to isolate viable CTCs from peripheral blood of
   metastatic patients, from which we have generated 2D and 3D in vitro
   (spheroids and organoids) and in vivo (CTC-derived xenografts) models.
   Particularly, drug screens on CTC-derived organoids were feasible within
   therapeutic timeframes which can potentially influence personalized treatment
   strategy. Drug responses from the screen mirrored patients’ drug resistance
   and revealed promising candidates for the treatment of individual patients.
   Beyond that, high-throughput drug screens in CTC-derived preclinical models
   closely mimicking patients' settings enable discovery, repurposing and
   development of more efficient cancer therapeutics.
   
   Integration of drug screening of liquid biopsy-derived tumor cells
   constitutes a powerful tool to better improve personalized treatment
   strategies and discovery for metastastic patients.

APPLICATIONS OF SINGLE CELL ANALYSIS TO DISEASE STUDIES

Session Chair: Kevin Eggan, Ph.D., Harvard University (USA)

 * Villages in a Dish: Scaling the use of human cell models to detect
   drug-genotype interactions
   Kevin Eggan, Harvard University
   
   A maturing application of reprogramming and stem cell technologies is their
   application to understanding how genetic variation that underlies disease
   risk impinge the function of affected cell types. However, a major limitation
   of this approach has been the number of patients and genetic variants that
   can be reasonably analyzed. I will describe a new strategy we have developed
   that allows us to simultaneously measure phenotypes in cell types derived
   from as many as 100 individuals in a single tissue culture well. These
   approaches, we call “Dropulation Genetics” and “Census sequencing” not only
   have allowed us to probe how genotype underlies phenotype at previously
   impractical scales, they have also provided a remarkable improvement in
   sensitivity and assay reproducibility. I will describe practical application
   of these approaches in psychiatry, neuromuscular disease and susceptibility
   to infectious agents.

 * Developing Epileptic Encephalopathy Models Using iPSC-Based Technologies
   Evangelos Kiskinis, Northwestern University Feinberg School of Medicine
   
   Mutations in KCNQ2, which encodes a pore-forming K+channel subunit
   responsible for neuronal M-current, cause neonatal epileptic encephalopathy,
   a complex disorder presenting with severe early-onset seizures and impaired
   neurodevelopment. The condition is exceptionally difficult to treat,
   partially because the effects of KCNQ2mutations on the development and
   function of human neurons are unknown. Here, we used induced pluripotent stem
   cells and gene editing to establish a disease model and measured the
   functional properties of patient-derived neurons using electrophysiological
   and optical approaches at single-cell resolution. We find that while
   patient-derived excitatory neurons exhibit reduced M-current early, they
   develop intrinsic and network hyperexcitability progressively. This
   hyperexcitability is associated with faster action potential repolarization,
   larger afterhyperpolarization, and a functional enhancement of Ca2+-activated
   K+(BK and SK) channels. These properties facilitate a burst-suppression
   firing pattern that is reminiscent of the interictal electroencephalography
   pattern in patients. Importantly, we were able to phenocopy these
   excitability features in control neurons only by chronic but not acute
   pharmacological inhibition of M-current. Our findings suggest that
   dyshomeostatic mechanisms compound KCNQ2 loss-of-function and lead to
   alterations in the neurodevelopmental trajectory of patient-derived neurons.
   Our work has therapeutic implications in explaining why KCNQ2 agonists are
   not beneficial unless started at an early disease stage.

 * Application of Microwell Plates in Single-Cell Analysis of T Cell-Mediated
   Tumor Cell Killing for High-Throughput Pharmacological Analyses
   Katherine Kozak, Genentech
   
   The ability to observe and quantitate T cell-mediated tumor cell killing at
   the individual cell level is critical for understanding the mechanism of
   immune activation and exhaustion to assist therapeutic designs. To acquire a
   large amount of single-cell data for statistical analysis, images of cells in
   micro-gridded chambers (sub-wells) within a standard microwell are captured
   to analyze single-cell interactions. The current available gridded platforms,
   manual microscopy imaging or standard automated image cytometer methods,
   however, are time-consuming. In addition, the analysis software is typically
   used for custom-made sub-wells constructed with Polydimethylsiloxane. In this
   work, we reported a high-throughput single T cell killing assay utilizing the
   Celigo Image Cytometer and Elplasia SQ plates that convert each 384-well into
   86,400 sub-wells. In this assay, both T cells and tumor cells were seeded
   with varying densities, resulting in 1 to 100 cells in each sub-well, as well
   as varying T cell activation reagent. The plate was scanned immediately after
   cell seeding at t = 0, 4, 22, 46, 68, and 168 hours, where the number of
   cells were tracked in each individual sub-well over time for each cell type.
   The image cytometer was able to rapidly acquire and analyze images at 1
   µm2/pixel. The images and results were exported into a custom program to
   determine the proper sub-well location of the segmented cells to allow
   separate tracking of small groups of tumor and T cells. This approach gave
   the image cytometry method greater capability in resolving subpopulations
   within the biological sample, resulting in more detail on the cytotoxic
   killing as a function of cell demographics in the tumor microenvironment.

 * Microfluidic Platform for Screening of Antibiotic Susceptibility at the
   Single-Cell Level
   Witold Postek, Institute of Physical Chemistry of the Polish Academy of
   Sciences
   
   The inoculum effect describes a dependency between the minimum inhibitory
   concentration (MIC) of an antibiotic and the concentration of bacteria in the
   sample: the less the bacteria, the less concentrated antibiotic is needed to
   stop their growth. MIC for populations consisting of a single cell is known
   as single-cell MIC (scMIC). scMIC is important for public health, as the
   presence of antibiotics at a concentration of scMIC in a large population of
   bacteria drives the evolutionary pressure towards resistant strains1, and the
   inoculum effect is a source of errors in MIC assessment in the clinic.
   However, efficient assessment of scMIC values for large numbers of cells has
   not been shown until now.
   
   Here, we demonstrate a method of determining scMIC values in hundreds of
   replications per experimental run, and we achieve this without optical
   labeling of the reaction conditions. We generate a series of emulsions of
   different concentrations of antibiotics at a step emulsifier2. We encapsulate
   single cells in each emulsion droplet due to stochastic confinement. Each
   emulsion is separated from the others by being encapsulated in a third
   immiscible phase and transferred to a piece of tubing, where all the
   separated emulsions can be incubated to provide for growth of bacteria. We
   measured the scMIC value of cefotaxime in E. coli for hundreds of cells,
   recording the inoculum effect when we used higher initial cell densities and
   observing the distribution of resistance level in a population of bacteria.
   Currently, we use our platform to generate up to 20 separate emulsions with
   different and known reaction conditions of ca. 2000 droplets each with
   immediate plans to upscale. In the near future we plan to screen for
   interactions of antibiotics in relation to inoculum effect, including the
   measurements at the single-cell level.
   
   The described method might be useful in the field of antibiotic resistance at
   a single-cell level, which is unbiased by the inoculum density. A
   microfluidic method of screening multiple chemical conditions in emulsions
   without labeling can be also deployed in other fields of research, wherever
   several reaction conditions should be replicated hundreds or thousands of
   times. For now3, to establish whether the bacteria grow or not, we detect
   fluorescence from fluorescent proteins produced by bacteria, but we are
   currently working on an add-on module to detect growth without labelling. We
   are also integrating our system with optical detection of moving droplets to
   automate the liquid handling protocol.
   
   1. T. Artemova, Y. Gerardin, C. Dudley, N. M. Vega and J. Gore, Mol. Syst.
      Biol., 2015, 11, 822.
   2. W. Postek, T. S. Kaminski and P. Garstecki, Lab Chip, 2017, 17, 1323–1331.
   3. W. Postek, P. Gargulinski, O. Scheler, T. S. Kaminski and P. Garstecki,
      Lab Chip, 2018, 18, 3668–3677.

SYSTEMATIC DATA-BASED APPROACHES FOR PRECISION MEDICINE

Session Chair: Kelly Frazer, Ph.D., University of California, San Diego 

 * Overlap of Fetal-Specific Cardiac Regulatory Variants and GWAS Lead Variants
   Supports Fetal Origins of Cardiovascular Disease
   Kelly Frazer, University of California, San Diego
   
   It has been hypothesized that many disease-causing variants exert their
   effects during development, rather than in adult cells. However, it is
   difficult to identify these variants and their effects as they could act in
   multiple different cell types, and there was a recent moratorium on research
   using fetal tissue. We recently established that iPSC-derived cardiovascular
   progenitor cells (CVPCs) are fetal-like, and can be utilized to identify
   cardiac regulatory variants. Here, we leveraged this system to identify fetal
   cell-type-specific eQTLs that underlie GWAS signals for adult cardiac
   diseases. We started by characterizing the differentiation of iPSCs into
   iPSC-CVPCs via scRNA-seq on eight samples, and found they were comprised of
   two cardiac cell types: cardiomyocytes (CMs) and epicardium derived cells
   (EPDCs). Next, we derived 180 iPSC-CVPCs, performed bulk RNA-seq, and used
   the scRNA-seq expression signatures to deconvolute and determine the relative
   proportions of CMs and EPDCs in each sample. We integrated these data with
   WGS and identified cell type-specific eQTLs (associated with only CMs or
   EPDCs). We next identified fetal-specific eQTLs by colocalizing our iPSC-CVPC
   eQTLs with all GTEx adult cardiac tissue eQTLs. To identify variants
   underlying the fetal origin of complex adult cardiac traits, we colocalized
   these fetal-specific eQTLs with cardiac traits GWAS summary statistics (pulse
   rate and myocardial infarction) and found 10 fetal-specific eGenes, including
   CLPTM1 which has previously been associated with congenital malformations (as
   expected for a fetal-acting gene). Our findings provide genetic evidence
   supporting the fetal origin of cardiovascular disease and show that
   iPSC-derived tissues can be leveraged to study the fetal origins of diseases
   in relevant cell-types.

 * Presentation Title TBD
   Paul de Bakker, Vertex

 * CURATE.AI-Enabled Personalised Dosing for Multiple Myeloma
   Agata Blasiak, National University of Singapore
   
   Standard of care therapy for various indications, including multiple myeloma
   (MM), is a combination of up to 4 drugs. Precision medicine has emerged as a
   game-changing approach for selecting drug combinations tailored to an
   individual to avoid treatment failure in a common case of drug resistance. To
   maximize the therapeutic outcome with the identified combination therapy, the
   drug dosing strategy should undergo an analogical approach - personalized
   dose selection for each drug tailored to an individual. Conventional
   approaches - titration, additive drug design, and dose escalation – often
   fail to find the optimal doses. Modern approaches - predictive algorithms and
   genotypic modeling – require a substantial amount of data and are costly. In
   this pilot study, we use CURATE.AI, a disease mechanism-independent and
   indication agnostic platform, to create an N-of-1 drug interaction profile
   using only the patient’s own data to identify optimized doses. CURATE.AI has
   been already clinically validated and has been used to optimize combination
   therapy for acute lymphoblastic leukemia, combination therapy for prostate
   cancer, liver transplant immunosuppression, and tuberculosis therapy, among
   other indications. We applied CURATE.AI to retrospectively analyze medical
   dataset in accordance with institutional IRB. As indicated in the medical
   records, a patient was given 14 monthly modulated dosages of revlimid and
   cyclophosphamide and a constant monthly dosage of dexamethasone. Quadratic
   polynomial correlation between the drugs’ dosages and the platelet count - a
   clinical indicator of the disease progression - was used to create the
   patient-specific CURATE.AI profile, which served as a map to identify drug
   dosages within clinically-accepted ranges that would result in an optimum
   platelet count. Using CURATE.AI analysis, the platelet count (P(r,c)) was
   correlated to revlimid and cyclophosphamide concentrations (r and c,
   respectively) by the following function:
   P(r,c)=62+4.690r-0.069rc+0.275r2+0.002c2, with R2value of 0.724 and a fitting
   correlation of 0.851. The CURATE.AI profile guided that to sustain the
   platelet count within the desired range (132-372x109/L), the patient should
   be given above 10 mg matched with c below 150 mg, or cabove180 mg matched
   with below 15 mg. CURATE.AI is deterministic and does not involve any
   prediction or uncertainty of response. In addition, CURATE.AI recommends
   doses within clinically-accepted ranges at patient-specific time points to
   optimize treatment response for that particular patient. The ability to
   identify patient-specific response constants has a paradigm-shifting
   potential – combining precision medicine for drug selection with CURATE.AI
   for dose selection brings us a step closer to the true realization of
   personalized medicine. We have also initiated a clinical trial that uses
   CURATE.AI for prospective dosing in MM (Clinicaltrials.gov: NCT03759093).

 * Personalizing Digital Therapeutics with CURATE.AI Identified N.1 Profiles
   Theodore Kee, National University of Singapore
   
   Digital therapeutics have emerged as an alternative or complementary modality
   of treatment to drug-based therapies for various indications, such as
   addiction and cognitive decline. Similar to conventional drug dosing, digital
   therapies often rely upon either fixed, or step-wise increased difficulty.
   Those approaches lack the flexibility and the ability to personalize the
   treatment to the individual. URATE.AI - a clinically validated deterministic
   optimization artificial intelligence (AI) platform that has already been used
   to modulate optimized dosing regimens for indications ranging from oncology
   (solid tumor/hematologic) to infectious diseases (HIV/TB) and
   immunosuppression (liver). In this prospective study, CURATE.AI identified
   individualized N-of-1 (N.1) learning trajectory profiles of healthy
   volunteers (both sexes, ages 21-40) trained on the Multi-Attribute Task
   Battery (MATB). MATB is a flight deck simulator developed by the National
   Aeronautics and Space Administration (NASA) and United States Air Force
   (USAF). The prospective clinical trial study design is a randomized,
   multiphase, parallel three-arm, single-blinded, N-of-1, single-center,
   exploratory pilot trial with 1:1:1 allocation approved by NUS Institutional
   Review Board (S-17-180) and listed under Clinicaltrials.gov identifier
   NCT03832101. For the CURATE.AI arm of the study, five subjects were fluent in
   English, had no prior experience with the MATB, and no history of perceptual
   or memory deficits, and recruited at Yale-NUS to participate in MATB
   simulator experiment sessions, conducted at the Yale-NUS campus. Each subject
   underwent a 34-minute MATB training session composed of 17 training and
   testing blocks of varying intensity levels (high, medium, low).
   Individualized CURATE.AI profiles were calibrated from the individual’s data:
   performance scores (RMAN-COMM z-scores from the training blocks), performance
   improvement, and training intensity levels. From each individual’s
   prospectively obtained data, N.1 learning trajectory profiles were derived
   and constructed with CURATE.AI, demonstrating the unique relationship between
   performance, training intensity, and performance improvement. Each subject
   had a different performance range (-1.24 to 0.70, -1.59 to 1.04, -2.13 to
   0.66) and different training intensity levels for optimal performance
   improvement. As identified from their CURATE.AI N.1 profiles, high-intensity
   training in select participants corresponded with greatest gains in
   performance improvement, while low-intensity training was identified for
   mediating similar gains in the other subjects. Based upon each individual’s
   unique interaction between performance and training intensity, these N.1
   learning trajectory profiles provide a means of real-time optimization of
   performance improvement by dynamically identifying and modulating the
   appropriate training intensities. In this prospective in-human study,
   interfacing MATB with CURATE.AI revealed substantial differences between
   subjects’ N.1 learning profiles and the correlation between tailored training
   intensity on performance improvement. The ability of CURATE.AI to identify
   N.1 profiles represents the advancement and utilization of AI to actionably
   address challenges encountered in personalized learning and the emerging
   field of digital therapeutics.

Back to Top

--------------------------------------------------------------------------------


OUR SPONSORS

Premier Sponsor:


 * Media Partner

 * Media Partner

 * Media Partner

 * Diamond Sponsor

 * Diamond Sponsor

 * Diamond Sponsor

 * Diamond Sponsor

 * Diamond Sponsor

 * Diamond Sponsor

 * Platinum Sponsor

 * Platinum Sponsor

 * Platinum Sponsors

 * Platinum Sponsor

 * Platinum Sponsor

 * Platinum Sponsor

 * Gold Sponsor

 * Gold Sponsor

 * Gold Sponsor

 * Gold Sponsor

 * Silver Sponsor

 * Silver Sponsor

 * Silver Sponsor

 * Silver Sponsor

 * Media Partner

 * Media Partner

 * Media Partner

 * Media Partner

 * Media Partner

 * Media Partner

 * Media Partner

 * Media Partner

 * Media Partner

 * Media Partner

 * Media Partner

 * Media Partner

 * Media Partner

 * Media Partner

 * Media Partner

 * Diamond Sponsor

 * Diamond Sponsor

 * Diamond Sponsor



SLAS HOME

Copyright © 2023
SLAS2020 International Conference & Exhibition
1301 West 22nd Street, Suite 708 • Oak Brook, IL 60523 USA
Telephone: +1.877.990.SLAS (7527)
Fax: +1.630.741.SLAS (7527)
Email: slas@slas.org   •   Privacy Notice   •   EU/UK Privacy Notice   •   SLAS
Home