Cambridge Healthtech Institute's 3rd Annual

PROTACs and Targeted Protein Degradation – Part 2

Proteomic Assays and Screening Strategies for Identifying Degradation Targets

September 29 - 30, 2021 EDT

The ubiquitin-proteasome system (UPS) and the autophagy-lysosome system are responsible for protein degradation and maintenance of proteostasis. They consist of well-controlled, selective mechanisms for intracellular protein degradation, however, the diversity and complexity makes it difficult to target these pathways for therapeutic intervention. Better understanding of ways to modulate these pathways using chemical probes, small molecule ligands and innovative screening platforms have made some UPS and autophagy targets more "druggable". Cambridge Healthtech Institute’s conference on PROTACs and Targeted Protein Degradation will bring together chemists, biologists and pharmacologists to discuss the progress that has been made and the challenges that are yet to be overcome. The UPS and autophagy pathways hold a lot of promise in seeking out previously “undruggable” targets for therapeutic intervention. The second part of the PROTACs and Targeted Protein Degradation conference will focus on emerging assays and screening strategies for identifying ligases, chaperone proteins, DUBs and other proteins that can be modulated for targeted protein degradation.

Wednesday, September 29

PLENARY KEYNOTE PROGRAM

11:30 am

Plenary Chairperson’s Remarks

An-Dinh Nguyen, Team Lead, Discovery on Target, Cambridge Healthtech Institute
Sunny Al-Shamma, President, Beacon Discovery a Eurofins Company
11:45 am

PLENARY: G Protein-Coupled Receptors and Beta Arrestin-Coupled Receptors: A Tale of Two Transducers

Robert J. Lefkowitz, MD, James B. Duke Professor of Medicine, Professor of Biochemistry, Duke University Medical Center; Investigator, Howard Hughes Medical Institute; 2012 Nobel Laureate in Chemistry

Beta arrestins are ubiquitous multifunctional adaptor proteins which mediate desensitization, endocytosis and signaling of most GPCRs. My lecture will cover how they were discovered as the mediators of rapid GPCR desensitization; the appreciation of their roles in endocytosis and, counterintuitively, as signal transducers in their own right; their roles in biased GPCR signaling and its therapeutic implications; and current understanding of the conformational basis of biased signaling.

12:20 pm LIVE:

Q&A Plenary Discussion

Panel Moderator:
Annette Gilchrist, PhD, Associate Professor, Pharmaceutical Sciences, Midwestern University
Panelist:
Robert J. Lefkowitz, MD, James B. Duke Professor of Medicine, Professor of Biochemistry, Duke University Medical Center; Investigator, Howard Hughes Medical Institute; 2012 Nobel Laureate in Chemistry
12:30 pm

PLENARY: Next-Generation Targeted Molecular Therapies

Alexandra Glucksmann, PhD, President & CEO, Cedilla Therapeutics

Despite decades of work, the need for small molecule-based targeted therapy in oncology is still immense. Amino-acid sequence and structure has been the primary lens to understand protein function, which has limited the reach of some key cancer targets. I highlight how we are accessing key cancer drivers that have been considered undruggable by considering the native full-length protein together with the relevant post-translational modifications, protein-protein interactions, and sub-cellular localization.

1:05 pm LIVE:

Q&A Plenary Discussion

Panel Moderator:
Joe Patel, PhD, Vice President, Structural Biology, Treeline Biosciences
Panelist:
Alexandra Glucksmann, PhD, President & CEO, Cedilla Therapeutics
1:15 pm Enjoy Lunch on Your Own
1:55 pm Refreshment Break in the Exhibit Hall with Poster Viewing

EMERGING STRATEGIES FOR PROTEIN DEGRADATION (VIRTUAL SESSION)

2:35 pm Organizer’s Welcome Remarks
Daniel Nomura, PhD, Professor of Chemistry, Molecular and Cell Biology, Nutritional Sciences and Toxicology, University of California, Berkeley

The Nomura Research Group is focused on reimagining druggability using chemoproteomic platforms to develop transformative medicines. One of the greatest challenges that we face in discovering new disease therapies is that most proteins are considered “undruggable,” in that most proteins do not possess known binding pockets or “ligandable hotspots” that small-molecules can bind to modulate protein function. Our research group addresses this challenge by advancing and applying chemoproteomic platforms to discover and pharmacologically target unique and novel ligandable hotspots for disease therapy. This talk will focus on using chemoproteomic platforms to expand the scope of targeted protein degradation technologies, and to develop new induced proximity-based therapeutic modalities. Collectively, our lab is focused on developing next-generation transformative medicines through pioneering innovative chemical technologies to overcome challenges in drug discovery.

3:15 pm

Strategies to Identify Molecular Glue Degraders at Scale

Cristina Mayor-Ruiz, PhD, Group Leader, Targeted Protein Degradation & Drug Discovery, Institute for Research in Biomedicine, Barcelona

Molecular glue degraders have incredible therapeutic potential, particularly considering delivering on the promise of degrading “undruggable” proteins. However, their discovery has thus far been largely serendipitous. We have established a scalable strategy toward glue degrader discovery that is based on chemical screening in cell models where many E3 ligases are functionally impaired. Collectively, we outlined a versatile and broadly applicable strategy to identify molecular glues at scale and thus empower future drug discovery efforts.

3:45 pm

A New Approach to Targeted Protein Degradation

Andrea Testa, PhD, Head, Chemistry, Amphista Therapeutics Ltd.

During the talk Amphista's approaches to Targeted Protein Degradation (TPD) will be discussed. In particular, I will be highlighting the need for new degrading mechanisms to increase the reach of TPD approaches even further. I will also be sharing new data on bifunctional degraders with improved profiles over PROTACs and discussing future directions for next-generation TPD drug discovery.

4:15 pm Refreshment Break in the Exhibit Hall with Poster Viewing
4:55 pm

FEATURED PRESENTATION: Detecting and Rebalancing Proteostasis Dysfunctions through Epichaperomes

Gabriela Chiosis, PhD, Member and Tri-Institutional Professor, Sloan Kettering Institute for Cancer Center, Cornell and The Rockefeller University

This talk will focus on detecting and rebalancing proteostasis dysfunctions through epichaperomes, maladaptive scaffolding platforms that alter protein-protein interaction networks in disease. Epichaperome detection and targeting in the context of diseases such as cancer and neurodegenerative disorders including Alzheimer’s will be presented.

5:25 pm

Protein Control Using Targeted Degradation Approaches

Behnam Nabet, PhD, Assistant Professor, Human Biology Division, Fred Hutchinson Cancer Research Center

Tag-based targeted protein degradation technologies including the degradation tag (dTAG) system have emerged as powerful approaches to control protein abundance using small molecule degraders. This talk will describe the development and advances of the dTAG technology platform and highlight case studies demonstrating utility for pre-clinical target discovery and validation.

5:55 pm

The Vital Role of Proteomics in Characterizing Protein Degraders

Fiona Pachl, PhD, Senior Scientist, Chemical Biology and Proteomics, AstraZeneca
Katelyn Cassidy, PhD, Senior Scientist, Chemical Biology, AstraZeneca

In recent years, the targeted protein degradation (TPD) field has developed at lighting speed. The Chemical Biology and Proteomics department at AstraZeneca has invested substantial resources into keeping up with the pace. In this talk, we will highlight the key capabilities that we have developed to meet the demands of our current TPD projects and to share how we aim to extend our impact in the near future.

6:25 pm Close of Day

Thursday, September 30

7:30 am Registration Open and Morning Coffee

UNDERSTANDING DEGRADATION PATHWAYS (VIRTUAL SESSION)

8:00 am

Exquisitely Specific Anti-KRAS Biodegraders Inform on the Cellular Prevalence of Nucleotide-Loaded States

Pooja Gopal, PhD, Senior Scientist, Quantitative Biosciences, MSD

Mutations to RAS are among the most common oncogenic drivers. To determine the advantages and feasibility of KRAS targeted degradation, we applied ‘Biodegraders’: chimeric proteins with a high-affinity target-binding domain fused to an engineered E3 ligase adapter. A series of anti-RAS biodegraders spanning different RAS isoform/nucleotide-state specificities and leveraging different E3 ligases provide evidence for KRAS degradability and its functional consequences. Furthermore, application of a GDP-state specific degrader uncovers the relative prevalence of the “off-state” of various KRAS mutants in the cellular context.

8:30 am

Cullin-RING E3 Ubiquitin Ligases (CRLs) for Targeted Protein Degradation 

Yue Xiong, PhD, Co-Founder & CSO, Cullgen

Recent advances in the development of small-molecule protein degraders, including molecular glues and heterobifunctional degraders, have made it possible to target many disease-causing proteins previously considered undruggable. The broad applicability of this approach is enabled by the plasticity of E3 ubiquitin ligases to interact with and ubiquitylate different (neo)substrates. The unique catalytic and assembly mechanism of the cullin-RING E3 ligases (CRLs) makes them particularly suitable for targeted protein degradation (TPD).

9:00 am

Use of Tip60 PROTACs in Cereblon-Knockin Mice

Wayne W. Hancock, MD, PhD, Professor of Pathology & Lab Medicine, University of Pennsylvania and Children's Hospital of Philadelphia

Finding new ways to target histone acetyltransferases, such as Tip60, is important for advances in immuno-oncology, and the PROTAC approach makes this possible. However, mice have a single amino acid substitution that blocks efficient iMID-dependent recruitment of the E3-ligase, Cereblon, limiting experimental studies. We report use of Tip60 PROTACs in WT vs. Cereblon knock-in mice in which PROTAC-dependent recruitment is now rendered active, allowing use of murine models for testing of this and other PROTAC molecules.

9:30 am Interactive Discussions

Interactive 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. For in-person events, the facilitator will lead from the front of the room while attendees remain seated. For virtual attendees, the format will be in an online networking platform. 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 website's Interactive Discussions page for a complete listing of topics and descriptions.

VIRTUAL INTERACTIVE DISCUSSION: Characterizing Protein Degraders

Fiona Pachl, PhD, Senior Scientist, Chemical Biology and Proteomics, AstraZeneca
Katelyn Cassidy, PhD, Senior Scientist, Chemical Biology, AstraZeneca
10:15 am Coffee Break in the Exhibit Hall with Poster Viewing

INDUCED PROXIMITY: PHARMACOLOGICAL CONSIDERATIONS (VIRTUAL SESSION)

11:00 am

Trivalent PROTACs: Enhancing Targeted Protein Degradation in Cancer Cells

Vesna Vetma, PhD, Cell Biologist, Department of Biological Chemistry and Drug Discovery, University of Dundee

PROTACs are required to form ternary complexes to drive target ubiquitination and degradation at low catalytic concentrations. This presentation will demonstrate a novel trivalent PROTAC strategy to boost targeted protein degradation. We will show how the trivalent degrader we discovered exhibits enhanced potency and downstream activity in a series of biophysical and cellular assays.

11:30 am

Extended Pharmacodynamic Response upon PROTAC-Induced Degradation of RIP2

Alina Mares, PhD, Senior Scientist, Protein Degradation, GlaxoSmithKline

PROTACs represents an exciting new modality in drug discovery that offers several potential advantages over traditional small-molecule inhibitors, including the potential to deliver pharmacodynamic (PD) efficacy which extends beyond the detectable pharmacokinetic (PK) presence of the PROTAC, driven by the synthesis rate of the protein. This disconnect between PK and PD, when coupled with low nanomolar potency, offers the potential for low human doses and infrequent dosing regimens with PROTAC medicines.

12:00 pm

Automated Analysis of Biphasic PROTAC Dose Response Data at Scale in Drug Development 

Ganesh Kadamur, Senior Research Scientist, AstraZeneca

Accurate SAR insights to drive drug development cycles depend on using the right mathematical models to fit data. For PROTACs data with hook behaviour, the standard Hill equation often misestimates pharmacological parameters including potency and efficacy. We have developed an automated, flexible bell-shaped biphasic model that can apply double sigmoidal curve fitting at scale and delivers accurate parameter estimates, enabling faster and more robust DMTA cycles.

Dengfeng Dou Dou, Vice President, Lead Generation Unit, HitGen Inc.

DNA encoded library technology is an optimal discovery tool for targeted protein degradation: 1) DEL screening is a very powerful way of identifying potent, selective POI and novel E3 ligase binders. 2) DEL molecule shares similar topological structure as PROTAC molecule with clearly defined linkage point; 3) it is proven that dual binding molecules can be directly identified from DEL screening; 4) DEL screening can be applied in molecular glue discovery.

12:45 pm Sponsored Presentation (Opportunity Available)
1:00 pm Session Break
1:00 pm Enjoy Lunch on Your Own
1:40 pm Refreshment Break in the Exhibit Hall with Last Chance Poster Viewing

INDUCED PROXIMITY: DESIGN CONSIDERATIONS (VIRTUAL SESSION)

2:15 pm

The Induced Proximity Platform at Amgen for Targeted Protein Degradation

Ryan Potts, PhD, Executive Director and Head, Induced Proximity Platform, Amgen, Inc.

Here I will discuss the latest targeted protein degradation developments from the Induced Proximity Platform at Amgen.


2:45 pm

Targeted Protein Degradation Using Ubiquitin Variant Induced Proximity

Wei Zhang, PhD, Assistant Professor, Department of Molecular and Cellular Biology, University of Guelph

Eukaryotic proteins undergo proteasomal degradation as a means of maintaining protein homeostasis, quantity, and quality to regulate various biological processes. An emerging theme in the drug development field is to exploit ubiquitin (Ub)-mediated proteolysis for targeted degradation of disease-causing proteins. We employ structure-based combinatorial protein design and engineering strategies to develop Ub variants (UbVs) as high-affinity binders for novel E3 ligases to induce intracellular degradation of selective substrates. 

3:15 pm

Structural Mechanisms of the Anaphase-Promoting Complex/Cyclosome (APC/C) E3 Ubiquitin Ligase

Nicholas G. Brown, PhD, Assistant Professor, Pharmacology, University of North Carolina at Chapel Hill

Ubiquitin (Ub)-dependent proteolysis drives the cell cycle. The behemoth anaphase-promoting complex/cyclosome (APC/C) is a 1.2 MDa Ub ligase responsible for regulating mitosis and G1 maintenance. Here, we will discuss how to optimize large complexes for cryo-EM studies using a new method called mass photometry and how multiple mechanisms activate the APC/C for a swift cell cycle transition.

3:45 pm Close of Conference