Cambridge Healthtech Institute’s 20th Annual

Proteomics-Driven Drug Discovery

Effective Use of Chemoproteomics, Chemical Biology, and Phenotypic Screening

September 26 - 27, 2023 EDT

While finding novel druggable targets and drug modalities for therapeutic intervention remains a top priority for the pharma/biotech industry, identifying and validating "good" targets and leads remains challenging. Cambridge Healthtech Institute’s conference on Proteomics-Driven Drug Discovery focuses on the use of mass spectrometry, biochemical, biophysical, cell-based assays, and AI/ML for target identification/deconvolution, lead discovery/optimization, and more. The conference brings together experts from academia and pharma/biotech to share ideas and case studies on how new chemical probes, proteomic assays, and screening platforms can be effectively used in drug discovery.

Tuesday, September 26

Registration and Morning Coffee7:00 am

Welcome Remarks7:55 am

COVALENT CHEMISTRIES & INDUCED PROXIMITY

8:00 am

Chairperson's Remarks

Daniel A. Erlanson, PhD, Senior Vice President, Innovation and Discovery, Frontier Medicines Corporation

8:05 am

Structural and Biochemical Characterization of a BTK-cIAP1 Covalent Complex

James Schiemer, PhD, Senior Scientist, Pfizer Inc.

Targeted protein degradation (TPD) using heterobifunctional chimeras holds the potential to expand target space and grow the ‘druggable proteome.'  Most acutely, this provides an opportunity to target proteins that lack enzymatic activity or have otherwise proven intractable to small molecule inhibition. Bridging covalent ligand discovery with chimeric degrader design has emerged as a potential mechanism to advance both fields.  Here, the authors, employ a set of biochemical and cellular tools to deconvolute the role of covalent modification in TPD using Bruton’s tyrosine kinase.  Results reveals that covalent target modification is fundamentally compatible with the protein degrader mechanism of action.

8:35 am

Leveraging Higher Throughput Targeted Proteomic Technologies to Impact PROTAC Portfolio

Uthpala Seneviratne, PhD, Associate Principal Scientist, AstraZeneca

Mass spectrometry-based targeted proteomics approaches have emerged as a promising technology to monitor absolute abundance of therapeutic target proteins that cannot be achieved via traditional protein detection technologies. We developed and applied these high-throughput strategies to address key MoA questions on PROTACs projects, including target protein and E3 abundance, protein turnover rates, and also for compound profiling. Case studies will be shown to demonstrate the technology and the impact on several PROTAC projects.

9:05 am

Discovery of Covalent Ligands Targeting CRL4DCAF2/DTL E3 Ligase for Targeted Protein Degradation

Kevin Webster, PhD, CSO, Frontier Medicines

Using our proprietary chemoproteomics platform, we identified site-selective covalent ligands for the CRL4DCAF2 E3 ligase. DCAF2 is a substrate receptor upregulated in specific cancers. A ligand was developed into hetero-bifunctional degraders that exhibited robust cellular DCAF2-dependent BRD2/4 degradation. Our work validates DCAF2 as a novel E3 ligase substrate receptor amenable to targeted protein degradation and demonstrates the utility of the Frontier Platform to discover novel ligands enabling proximity-inducing therapeutics.

Networking Coffee Break9:35 am

10:00 am

Chairperson's Remarks

Gizem Akcay, PhD, Head, Chemical Biology, Bayer Research and Innovation Center Cambridge

10:05 am

Identification of Small Molecule Covalent Inhibitors Disrupting YAP-TEAD Protein-Protein Interaction and Targeting Cancer Cells

Ramesh Kumar, PhD, Principal Investigator & Scientist, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR)

We report covalent small molecule inhibitors that form a covalent complex with a cysteine in the TEAD palmitoylation site. Compounds inhibited TEAD transcriptional target genes in cancer cells. Cellular proliferation and colony forming assay data strongly displayed cellular sensitivity hippo signaling altered cancer cell lines. Transcriptional profiling and RNA seq data indicate that the covalent inhibitors specifically downregulating the hippo signaling.

10:35 am

Advancing Covalent Ligand Discovery for Precision Oncology Targets

Heather Murrey, PhD, Senior Director, Scorpion Therapeutics

Recent chemoproteomics advances have enabled covalent ligand discovery across a broad range of new targets. Here, we discuss the expanding role of chemical biology and chemoproteomics to support covalent lead discovery efforts, from early hit-finding to late lead optimization.

11:05 am

Exploring Cell-Surface Protein Interaction Networks to Identify Novel Therapeutic Targets and Modalities

Noah Dephoure, PhD, Head of Proteomics, InduPro, Inc.

Cell-surface proteins present unique opportunities for diagnostic and therapeutic targeting. Despite major advances in proteomic methods, our understanding of surfaceome content and organization remains limited. Using photochemical proximity labeling, we are generating high-confidence protein interaction networks for cell surface proteins in diverse cell types and contexts. This data forms the centerpiece of an integrated Membrane Interactome knowledge-base (MInt) incorporating in-house generated data for target discovery and prioritization.

11:35 am Drug Discovery at the Melting point: Exploring and Validating Target Space with CETSA

Michael Dabrowski, CEO, Pelago Bioscience

The prerequisite of any biological effect is a drug-target interaction, which can be studied in live cells using the CETSA technology. As therapeutic target space expands to include harder to drug targets like non-enzymatic proteins the importance of target validation and understanding the MoA also increase. Because the CETSA technology is fully label-free, new targets and modalities can be studied in a physiological relevant format, illustrated through a series of use-cases.

 

Enjoy Lunch on Your Own11:50 am

LEVERAGING PROTEOMICS IN DRUG DISCOVERY

1:15 pm

Chairperson's Remarks

Christopher am Ende, PhD, Associate Research Fellow, Internal Medicine Medicinal Chemistry, Pfizer Inc.

1:20 pm

FEATURED PRESENTATION: Incorporating Targeted Approaches into Cysteine-Based Profiling Strategies

Steve Gygi, PhD, Professor, Department of Cell Biology, Harvard Medical School

Mass spectrometry is the enabling tool in chemoproteomics, and screening entire libraries of electrophilic compounds is now possible. In this talk, I will highlight our progress in generating targeted methods that allow profiling of up to 500 reactive cysteine sites in a single analysis.  Advantages include near complete datasets with fewer missing values and dramatically increased sensitivity. We demonstrate the approach screening a library in K562 cells in 384-well plates. 

1:50 pm

Harnessing Biomolecular Condensates: Therapeutic Potential of Condensate Modifying Drugs

Ann Boija, PhD, Vice President, Head of Cancer Biology, Dewpoint Therapeutics

This presentation will explore the innovative concept of developing condensate modifying drugs (c-mods) to address a variety of health conditions, from cancer to neurodegeneration. By altering the formation, stability, dynamics, and dissolution of biomolecular condensates, these novel therapies could pave the way for innovative solutions to previously "undruggable" targets, presenting groundbreaking opportunities for disease treatment.

2:20 pm

Analysis of Post-Translational Modifications from Proteomic Studies

Stephanie Ashenden, PhD, Senior Informatician, Artificial Intelligence & Machine Learning, AstraZeneca

Post-Translational Modifications (PTMs) are important chemical changes to a protein that can result in structural, functional and stability changes to the protein which, therefore, make them critical in a wide range of biological pathways. Common PTMs include phosphorylation, acetylation, and methylation. In this talk we will explore some of the known complexities with analysing PTMs and ways to support better biological interpretation around changes in a peptide sequence given a stimulus in comparison to another stimuli (or absence of one). We will also explore a downstream pipeline that allows us to analyse and interpret data. This will involve considerations around quality control, experimental design, normalisation, missing data and trends in data, differential expression and downstream analyses.

In-Person Group Discussions2:50 pm

In-Person Group Discussions are informal, moderated discussions, allowing participants to exchange ideas and experiences and develop future collaborations around a focused topic. Each discussion will be led by a facilitator who keeps the discussion on track and the group engaged. To get the most out of this format, please come prepared to share examples from your work, be a part of a collective, problem-solving session, and participate in active idea sharing. Please visit the In-Person Group Discussions page on the conference website for a complete listing of topics and descriptions.

IN-PERSON GROUP DISCUSSION 2A:

Leveraging Phenotypic Screening, Chemical Biology, and Chemoproteomics Tools

Gizem Akcay, PhD, Head, Chemical Biology, Bayer Research and Innovation Center Cambridge

Christopher am Ende, PhD, Associate Research Fellow, Internal Medicine Medicinal Chemistry, Pfizer Inc.

Steve Gygi, PhD, Professor, Department of Cell Biology, Harvard Medical School

Andrew Zhang, PhD, Director, Chemical Biology, AstraZeneca

  • How can proteomics be best used in the drug discovery pipeline?
  • Can traditional chemoproteomics and orthogonal methods help deconvolute “undruggable” targets?
  • Which improvements are most needed in developing good proteomic assays and screening platforms?
  • Insights into some of the new chemistries and chemical probes being generated​

Grand Opening Refreshment Break in the Exhibit Hall with Poster Viewing3:35 pm

4:15 pm

Advancing Chemical Biology through Innovative Chemistry 

Christopher am Ende, PhD, Associate Research Fellow, Internal Medicine Medicinal Chemistry, Pfizer Inc.

Chemical biology has broad influence on many drug discovery programs. For example, chemical biology tools and techniques can be used to understand target engagement, selectivity/off-target profiles and binding sites of lead matter, which can be critical to the progression of a project. This presentation will focus on the development of new and innovative chemistry to offer improved methods to access and use chemical probes in a variety of applications.

4:45 pm

Chemical Biology Approaches to Expand Druggable Proteome

Gizem Akcay, PhD, Head, Chemical Biology, Bayer Research and Innovation Center Cambridge

Targeted covalent inhibition of disease associated proteins has become a powerful methodology in drug discovery with the approval of commercial drugs for targets previously deemed undruggable. We will showcase examples of combining chemoproteomics approaches together with protein structure-function relation at residue level for identifying actionable targets and novel druggable pockets.

5:15 pm

Exposing and Exploiting Druggable Protein Landscapes with In Situ Proximity Barcoding Platforms

Raymond Moellering, PhD, Associate Professor, Department of Chemistry, University of Chicago

There are significant expanses of the potential druggable protein landscape that are relatively unexplored for small molecule drug discovery. In this talk, I will discuss the development and deployment of complementary in situ proximity barcoding chemoproteomic approaches to map protein activity states and/or interaction partners directly in native cellular environments. In particular, I will focus on the application of these platforms for functional ligand binding site discovery, high-throughput ligand profiling and mechanism-of-action interrogation for challenging protein classes.

Welcome Reception in the Exhibit Hall with Poster Viewing5:45 pm

Close of Day6:45 pm

Wednesday, September 27

Registration and Morning Coffee7:30 am

PROTEOMICS FOR TARGET DISCOVERY

7:55 am

Chairperson's Remarks

Doug Johnson, PhD, Senior Director, Chemical Biology & Proteomics, Biogen

8:00 am

Use of Reverse Phase Protein Arrays for Lead Optimization and Drug Target Activation Mapping 

Emanuel Petricoin, PhD, Professor & Co-Director, Center for Applied Proteomics & Molecular Medicine, George Mason University

Analysis of on and off-target effects during lead optimization, target validation, uncovering mechanisms of resistance and potential for rational  therapeutic combinations or sequencing, as well as drug target biochemistry and modulation are critical elements of the biopharmaceutical late stage preclinical pipeline. The reverse phase protein array technology is a powerful analytical platform that is able to quantitatively measure hundreds of protein drug targets and phosphorylated signaling architecture at once from microscopic quantities of cells and is especially well-suited for these efforts.

8:30 am Expression & Characterization of Novel GCN5-Related N-Acetyltransferases Enabled by Nuclera’s Protein Access System

Michael Chen, CEO, Nuclera

Hans Gerstmans, PhD, MasscheleinLab-Laboratory for Biomolecular Discovery & Engineering, MeBioS-Biosensors group (KU Leuven)

GCN5-related N-acetyltransferases (GNATs) are a family of enzymes that acetylate various molecules, including proteins and small molecules. They are part of the enzymatic machinery that bacteria use to modify antibiotics, resulting in antibiotic resistance. Despite the importance of these enzymes, they have not been obtained in our lab or reported in literature. Using Nuclera’s eProtein Discovery system, we found conditions to obtain active recombinant GCN5-related N-acetyltransferases.

9:00 am

Comparative Label-Free Proteomics Aiming at the Cell Surface Proteome of RAS-Driven Human Cancer Cells to Uncover Novel Druggable Targets

Josip Blonder, MD, Scientist Emeritus, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research

Ras isoform mutants play a major role in most aggressive and deadly cancers in humans (e.g., pancreas, lung, ovary). While the majority of drug discovery efforts target oncogenic Ras signaling inside the cell, only few a endeavors have been focused on the cell-surface. Using quantitative label-free membrane proteomics we were able to identify more than 500 cell surface proteins found unique or upregulated on the surface of KRasG12V -expressing cells.

9:30 am

An Aptamer Affinity-Based Proteomic Profiling Platform to Identify Modality Agnostic Novel Drug Targets Directly from Primary Patient Tissue

Heather O'Neill, PhD, Vice President, Precision Medicine, Target & Drug Discovery, Caris Discovery

Affinity purification-mass spectrometry (AP-MS) with aptamer libraries enriched directly on molecularly profiled cancer patient tissue was used for novel drug target discovery. This method allows for unbiased detection of cancer-specific proteins that elude classical proteomic identification methods. Targets were validated with RNA expression data from our clinico-genomic database and IHC on patient tissue. Cell surface targets were validated on cell lines by mass spectrometry, flow cytometry, immunocytochemistry, and with cytotoxicity assays.

Coffee Break in the Exhibit Hall with Poster Viewing10:00 am

PLENARY KEYNOTE PROGRAM

10:40 am

Plenary Chairperson’s Remarks

An-Dinh Nguyen, Team Lead, Discovery on Target, Cambridge Healthtech Institute

10:45 am

PLENARY: The New Science of Therapeutics

Jay E. Bradner, MD, Physician Scientist, Former President, Novartis Institutes for BioMedical Research, Inc.

I will share reflections on how new paradigms in the science of therapeutics are creating opportunities to approach historic challenges in medicine. Specifically, I will share approaches to targeting transcription factors and discuss how modularity is a paradigm for next-generation low-molecular weight and biological therapeutics. Finally, I will offer reflections on drug development and the fitness, opportunities, and challenges of the biomedical ecosystem.

11:30 am

PLENARY: Accelerating Drug Discovery Using Machine Learning and Cell Painting Images

Anne E. Carpenter, PhD, Senior Director, Imaging Platform & Institute Scientist, Broad Institute

Shantanu Singh, PhD, Senior Group Leader, Machine Learning, Imaging Platform, Broad Institute

Microscopy images can reveal whether a cell is diseased, is responding to a drug treatment, or whether a pathway has been disrupted by a genetic mutation. In a strategy called image-based profiling, often using the Cell Painting assay, we extract hundreds of features of cells from images. Just like transcriptional profiling, the similarities and differences in the patterns of extracted features reveal connections among diseases, drugs, and genes.

Close of Proteomics-Driven Drug Discovery Conference12:15 pm