Cambridge Healthtech Institute’s 4th Annual

PROTACs and Molecular Glues – Part 2

Covalent Modifications Inducing Proximity & Degradation

October 19 - 20, 2022 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 and hold a lot of promise in seeking out previously “undruggable” targets for therapeutic intervention. However, their diversity and complexity make it difficult to target these pathways for therapeutic intervention. Cambridge Healthtech Institute’s conference on PROTACs and Molecular Glues discusses the progress that has been made and the challenges that are yet to be overcome. The second part of the PROTACs and Molecular Glues conference will focus on emerging assays and screening strategies for identifying ligases and proteins that can be modulated for protein degradation. The use of novel covalent chemistries for inducing chemical proximity and use of AI/ML modeling to enable targeted protein degradation will also be discussed.

Wednesday, October 19

PLENARY KEYNOTE PROGRAM

ROOM LOCATION: Constitution A + B

11:00 am

Plenary Chairperson’s Remarks

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

11:05 am

PLENARY: Pirating Biology to Detect and Degrade Extracellular Proteins

James A. Wells, PhD, Professor, Departments of Pharmaceutical Chemistry and Cellular & Molecular Pharmacology, University of California, San Francisco

In contrast to intracellular PROTACs, approaches to degrade extracellular proteins are just emerging. I’ll describe our recent progress to harness natural mechanisms such as transmembrane E3 ligases to degrade extracellular proteins using fully genetically encoded bispecific antibodies we call AbTACs. We have also engineered a peptide ligase which can be tethered to cells to detect proteolysis events and target them with recombinant antibodies for greater selectivity for the tumor microenvironment.

11:50 am

PLENARY: Therapeutic Modalities for Neuroscience Diseases

Anabella Villalobos, PhD, Senior Vice President, Biotherapeutics & Medicinal Sciences, Biogen

Many effective medicines exist to treat neurological diseases, but medical need remains high. We have a unique multi-modality approach to discover novel therapies and our goal is to find the best modality regardless of biological target. With a multi-modality approach, we aim to expand target space, leverage synergies across modalities, and offer options to patients. Opportunities and challenges associated with small molecules, biologics, oligonucleotides, and gene therapy will be discussed.

Enjoy Lunch on Your Own12:35 pm

Refreshment Break in the Exhibit Hall with Poster Viewing (Grand Ballroom Foyer)1:25 pm

ROOM LOCATION: Constitution A

AI/ML FOR PROTEIN DEGRADATION

2:35 pmWelcome Remarks
2:40 pm

Chairperson's Remarks

Woody Sherman, PhD, CEO, Psivant Therapeutics

2:45 pm

Accelerating Rational Degrader Design via Computational Prediction of Ternary Structure Ensembles

Woody Sherman, PhD, CEO, Psivant Therapeutics

We describe a novel method that combines experimental biophysical data (HDX-MS) with weighted ensemble simulations (WES) to accurately predict ternary complex structures at atomic resolution. We show that the WES+HDX approach generates accurate structures (RMSD below 2.0 Å to x-ray) and can reproduce solution-state dynamic behavior of the ternary complex. We illustrate how this approach is used to predict degradation propensity of different heterobifunctional and glue molecules.

3:15 pm How Artificial Intelligence Enhances Drug Discovery

Sang Eun Jee, PhD, Application Scientist, Xtalpi

AI can cut down the development timeline and cost for drug discovery by answering two significant questions: What molecules should be made next and how are the lead molecules modified? AI technology in drug discovery will be introduced with case studies of how we solved challenging problems with AI.  The key to success in AI-driven drug discovery in the future will also be discussed with the lessons learned from history.

Dessert Break in the Exhibit Hall with Poster Viewing (Grand Ballroom)3:45 pm

4:25 pm

Estimating Target Degradability from Protein-Intrinsic Features

Shourya Roy Burman, PhD, Research Fellow, Cancer Biology, Dana-Farber Cancer Institute

Chemo-proteomics profiling of PROTACs designed from pan-class inhibitors revealed a large difference in the degradation frequencies of the target proteins engaged by these molecules. Using protein-intrinsic features, we developed a machine learning classifier that discriminates target proteins based on their observed degradation patterns and highlights properties that dictate their degradability. Using computational structural modeling, we provide mechanistic insight into the predicted features and obtain actionable information for rational PROTAC design.

4:55 pm

Closing the Gap: Heterogeneous Molecular Modeling & Machine Learning for Accurate Modeling

Victor Guallar, PhD, Professor, Barcelona Supercomputing Center and Nostrum Biodiscovery

Combining the state-of-the-art molecular modeling, in heterogeneous data sources and in machine learning techniques, we are dramatically increasing the accuracy in our computational predictions. We will showcase recent successful case studies including virtual screening enrichment, ligase screening for TPD and ternary complex formation in PROTACs. Overall, the enrichment of machine learning techniques with data augmentation from molecular modeling seems to provide the necessary boost that prediction models might need.

5:25 pm

PANEL DISCUSSION: Challenges with Using AI Predictions for Designing Protein Degraders

PANEL MODERATOR:

Woody Sherman, PhD, CEO, Psivant Therapeutics

PANELISTS:

Shourya Roy Burman, PhD, Research Fellow, Cancer Biology, Dana-Farber Cancer Institute

Victor Guallar, PhD, Professor, Barcelona Supercomputing Center and Nostrum Biodiscovery

Dinner Short Course Registration*5:55 pm

*Premium Pricing or separate registration required. See Short Courses page for details.

Close of Day9:00 pm

Thursday, October 20

Registration and Morning Coffee (Grand Ballroom Foyer)7:30 am

NEW LIGASES & DEGRADATION STRATEGIES

ROOM LOCATION: Constitution A

7:55 am

Chairperson's Remarks

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

8:00 am

Molecular Glues for Protein Degradation and Beyond

Eric Fischer, PhD, Associate Professor, Biological Chemistry and Molecular Pharmacology, Harvard Medical School; Director Center for Protein Degradation, Dana-Farber Cancer Institute

Small molecules that induce protein degradation through ligase-mediated ubiquitination, are showing considerable promise as a new pharmacological modality. Following clinical proof of concept by Thalidomide and related IMiDs, several new molecular entities are entering clinical development. Significant progress has been made towards chemically induced targeted protein degradation using heterobifunctional small molecule ligands and exciting opportunities arise from better understanding molecular glues. We will present recent work towards a better understanding of the molecular principles that govern neo-substrate recruitment by small molecule degraders.

8:30 am

Discovery of Novel E3 Ligands for Targeted Protein Degradation

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

Three distinct characteristics of targeted protein degradation empower drug discovery; the catalytic nature that can achieve high efficacy, the ability to target previously undruggable proteins and to deliver the drug activity to selective tissues. E3 ligands hold the key to realize the full potential of TPD, but are very limited at present. Cullgen has discovered multiple novel E3 ligands for the targeted protein degradation.

9:00 am

FEATURED PRESENTATION: An E3 Ligase Guide to the Galaxy of Small Molecule-Induced Protein Degradation

Michael Rape, PhD, Professor, Department of Cell & Developmental Biology, University of California, Berkeley; Investigator, Howard Hughes Medical Institute

Human cells express ~600 E3 ligases that in theory could be harnessed by small molecules to alter the stability or activity of pathological proteins. How to select an E3 ligase for therapeutic application is, however, still unclear. We have used biology to guide ligase selection: by discovering new pathways for ubiquitin-dependent protein quality control, we have unlocked new enzymes for targeted protein degradation. I will describe our recent insight and discuss possibilities of this mechanism-inspired approach to E3 ligase selection.

Interactive Discussions9:30 am

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. 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 conference website's Interactive Discussions page for a complete listing of topics and descriptions.

IN-PERSON INTERACTIVE DISCUSSION:

Targeted Protein Degradation – Where Are We Headed?

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

Nikki R. Kong, PhD, Senior Scientist; Biology Group Leader, Center for Protein Degradation, Dana-Farber Cancer Institute

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

  • Exploring novel E3 ligases and ligands for targeted protein degradation
  • Rational design strategies for molecular glue degraders
  • New assays and screening approaches for mechanistic studies of degraders and glues​

Coffee Break in the Exhibit Hall with Poster Viewing (Grand Ballroom)10:15 am

11:00 am

Target Protein Localization and Its Impact on PROTAC-Mediated Degradation

Gopal Sapkota, PhD, Programme Leader, MRC Protein Phosphorylation & Ubiquitylation Unit, Sir James Black Centre, School of Life Sciences, University of Dundee

Unique subcellular environment due to factors such as the location of the POI and access to the E3 ligase being recruited potentially impacts PROTAC efficacy. To interrogate whether subcellular context of POI influences PROTAC-mediated degradation, we expressed either Halo or FKBP12F36V (dTAG) constructs consisting of varying localisation signals and tested the efficacy of their degradation by von Hippel-Lindau (VHL)- or cereblon (CRBN)-recruiting PROTACs targeting either Halo or dTAG. POIs were localised to the nucleus, cytoplasm, outer mitochondrial membrane, endoplasmic reticulum, Golgi, peroxisome, or lysosome. Differentially localised Halo or FKBP12F36V proteins displayed varying levels of degradation using the same respective PROTACs.

11:30 am

Employing Phenotypic Assays to Dissect Mechanisms of Molecular Glue Degraders 

Nikki R. Kong, PhD, Senior Scientist; Biology Group Leader, Center for Protein Degradation, Dana-Farber Cancer Institute

Despite identification of IMiD-induced neo-substrates of E3 ligase CRL4-Cereblon in hematopoietic malignancies, the cell-type specific mechanisms of these molecules have not been extensively explored, especially in the immune context. As a result, there are a number of unknowns regarding the efficacy and safety of TPD therapeutics derived from these molecules. Here, we employed complex phenotypic assays to better understand the on-target mechanisms of this class of drugs. 

12:00 pm

Surveying the Landscape of Drug Resistance Mutations within Neosubstrates to Molecular Glue Degraders Using CRISPR Scanning

Brian Liau, PhD, Associate Professor, Department of Chemistry and Chemical Biology, Harvard University

CRISPR scanning was used to broadly survey the landscape of drug resistance mutations to molecular glue degraders in neosubstrates. Integrative analysis of resistance sites revealed varying levels of sequence conservation and mutational constraint that control the emergence of different resistance mechanisms, highlighting that many regions co-opted in targeted protein degradation are inessential. Altogether, we outline a rapid and general approach to survey neosubstrate requirements necessary for effective degradation.

12:30 pm Targeted Protein Degrader Discovery Using SPRINTer Cell-Based Assay

Chao Tsung Yang, PhD, Principal Scientist Group Leader, R&D Department, Eurofins DiscoverX

Rapidly screen small molecule therapeutics, evaluate target engagement, and measure changes in endogenous protein levels using SPRINTer functional, cell-based assays for multiple targets.

In this talk, we will discuss:

  • SPRINTer assay principle and characterization of bi-functional degraders and molecular glues for multiple targets
  • Assay applicability to evaluate protein stabilization using the SPRINTer CDKN1A (p21) assay to monitor TP53 modulation by MDM2 inhibitors
  • Evaluation of compound-target engagement using select SPRINTer cell lines

Enjoy Lunch on Your Own1:00 pm

Refreshment Break in the Hall with Poster Viewing (Grand Ballroom)1:40 pm

DEGRADATION APPROACHES FOR ONCOLOGY

2:10 pm

Chairperson's Remarks

Jun Yang, PhD, Professor and Vice Chair, Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital

2:15 pm

Therapeutic Evaluation of Multi-Targeted SRC Kinase PROTAC in Cancer

Jun Yang, PhD, Professor and Vice Chair, Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital

Clinically used as an ABL inhibitor, dasatinib promiscuously targets other tyrosine kinases, especially those in the SRC family. We developed a series of dasatinib-based PROTAC molecules, using phenyl-glutarimide as the cereblon binder. These SRC-directed PROTACs exhibited remarkable activity in LCK-activated T cell leukemia, in vitro, and in vivo. Because SRC family kinases are important therapeutic targets in cancer, we are evaluating these PROTACs systematically in ~1,000 tumors across histologic types.

2:45 pm

New Opportunities for Studying the Function of the Nucleosome Remodeling Factor, NURF through Inhibition and Degradation

William Pomerantz, PhD, Associate Professor, Department of Medicinal Chemistry, University of Minnesota, Twin Cities

Protein members of nucleosome remodeling complexes are emerging therapeutic targets. One protein of interest, the Bromodomain PHD Finger Transcription Factor, BPTF, is an essential member of the human nucleosome remodeling factor NURF. In recent years years, BPTF has become increasingly identified as a pro-tumorigenic factor, prompting investigations into the molecular mechanisms associated with BPTF function. Despite a druggable bromodomain small molecule discovery is at an early stage. Our lab has developed novel screening approaches including protein-based 19F NMR to develop both the first inhibitor of the BPTF bromodomain, and now more potent and selective chemical probes. Building on these results I will present our efforts at developing the first BPTF degraders to study the role of this protein in pediatric cancers.  

3:15 pm

Positive Selection Screens for Oncoprotein Degraders

Vidyasagar Koduri, MD, PhD, Instructor in Medicine, Harvard Medical School; Department of Hematology, Brigham and Women’s Hospital

Matthew Oser, MD, PhD, Assistant Professor, Department of Medicine, Dana-Farber Cancer Institute, Harvard Medical School

Current loss of signal ("down") assays for identifying degraders often exhibit poor signal-to-noise ratios and false positives from compounds that nonspecifically suppress transcription or translation. Here we describe a gain of signal ("up") assay for degraders that can be used in both chemical and genetic screens. This should facilitate the identification of drugs that directly or indirectly degrade undruggable proteins.

Close of Conference3:45 pm