Cambridge Healthtech Institute's 3rd Annual

PROTACs and Targeted Protein Degradation – Part 1

Molecular Glues, Chimeras and Covalent Inhibitors as Novel Degraders

September 28 - 29, 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 first part of the PROTACs and Targeted Protein Degradation conference will discuss ways to design and optimize small molecule degraders such as, proteolysis-targeting chimeras (PROTACs), E3 ligase and deubiquitinating enzyme (DUB) modulators, thalidomides and other molecular glues. Challenges that exist in terms of stability, biodistribution and penetration of these molecules for better in vitro to in vivo translation will be discussed.

Monday, September 27

11:30 am Virtual Short Courses

Please visit the Short Courses page for details. Premium Package or separate registration required.

Tuesday, September 28

7:00 am Registration Open and Morning Coffee

UNDERSTANDING MECHANISMS FOR BETTER DESIGN OF DEGRADERS

7:55 am Organizer’s Welcome Remarks
8:00 am

Chairperson's Remarks

Daniel A. Erlanson, PhD, Vice President, Chemistry, Frontier Medicines Corporation
8:05 am

Snapshots and Ensembles of BTK and cIAP1 Protein Degrader Ternary Complexes

Matthew Calabrese, PhD, Senior Director & Head, Structural & Molecular Sciences, Pfizer Global R&D Groton Labs

Heterobifunctional chimeric degraders are a class of ligands that recruit target proteins to E3 ubiquitin ligases to drive compound-dependent protein degradation. Critical to the mechanism of action is the formation of a ternary complex between the target, degrader and E3 ligase to promote ubiquitination and subsequent degradation. However, limited insights into ternary complex structures exist, including a near absence of studies on one of the most widely co-opted E3s, cellular inhibitor of apoptosis 1 (cIAP1). Our results reveal insights from unique ternary complex structures and show that increased ternary complex stability/rigidity need not always correlate with increased degradation efficiency.

8:35 am

Small-Molecule Induced Polymerization as Novel Mechanism for Protein Degradation Therapeutics

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, have shown considerable promise as a new pharmacological modality. Thalidomide and related IMiDs provided the clinical proof of concept, while significant progress has recently been made towards chemically induced targeted protein degradation using heterobifunctional small molecule ligands. We will present recent work towards a better understanding of the molecular principles that govern neo-substrate recruitment, and other small molecule degraders.

9:05 am

Tumor-Targeted Protein Degradation via Chaperones

Kevin Foley, PhD, Co-Founder & CSO, Ranok Therapeutics

Cellular chaperones not only mediate protein folding, but can also direct proteins towards the ubiquitin-proteasome system. Chaperone-mediated protein degraders (CHAMPs) are hetero-bifunctional small molecules that induce proximity between a target of interest and the chaperone machinery, thereby resulting in target degradation. Due to the highly activated state of chaperones in cancer cells, CHAMPs preferentially induce protein degradation in tumors relative to normal tissues.

9:35 am Coffee Break in the Exhibit Hall with Poster Viewing

DESIGN & DEVELOPMENT OF NOVEL PROTEIN DEGRADERS

10:25 am

Targeting Membrane Proteins with Novel PROTAC Drugs

Tauseef Butt, PhD, President & CEO, Progenra, Inc.

Currently, PROTACs based on Cereblon, VHL, HDM2 and cIAPs degrade a limited set of therapeutic targets. Targeting membrane protein for degradation can open new applications for PROTAC drugs that will compete with monoclonal antibody therapies and yet have an advantage of small molecules. Progenra has established a novel membrane associated ubiquitin ligase that preferentially degrades membrane proteins. Importance of this ligase in selective regulation of membrane receptor provides new ways to regulate membrane receptor function with small molecules. This presentation will provide examples of PROTAC mediated degradation of membrane receptors.

10:55 am

A Rationally Designed BCL-XL and BCL-2 Dual Degrader with Improved Anti-Leukemia Activity

Daohong Zhou, MD, Professor, Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville

PROTACs have emerged as a novel drug development platform. Through computational modelling of the entire NEDD8-VHL Cullin RING E3 ubiquitin ligase (CRLVHL)/PROTAC/BCL-xL/UbcH5B(E2)-Ub/RBX1 complex, we rationally designed of a new series of ABT263-derived and VHL-recruiting PROTACs and generated the first potent BCL-xL and BCL-2 (BCL-xL/2) dual degrader with significantly improved antitumor activity against BCL-xL/2-dependent leukemia cells. In addition, our studies provide the first experimental evidence that the accessibility of lysine(s) on a target protein plays an important role in determining the selectivity and potency of a PROTAC in inducing protein degradation.

11:25 am

Discovery of Oral PROTACs Targeting AR for Prostate Cancer

Wu Du, PhD, Senior Vice President, Department of Medicinal Chemistry, Hinova Pharmaceuticals, Inc.

Prostate cancer remains as an unmet medical need despite that there are androgen receptor (AR) inhibitors on market. PROTAC-targeting AR signaling emerges as a showcase of this revolutionary strategy, and a new approach to treat prostate cancers. Recent progress in oral PROTAC for prostate cancer from Hinova will be presented.


Ksenya Cohen Katsenelson, PhD, Group Leader, R&D Department, Eurofins Discovery
Chao-Tsung Yang, PhD, Principal Scientist, Eurofins DiscoverX

Eurofins Discovery is diversifying its novel E3scan ligand-binding platform with additional E3 ligase target assays and SPRINTer cell lines for new disease targets. Here, we present E3scan validation data for E3 ligases that have not been utilized in targeted protein degradation yet. In addition, we will demonstrate how new SPRINTer cell lines aim to identify degraders for BTK tyrosine kinase and modulators for the MDM2/p53 E3 ligase complex.

12:25 pm Session Break
Erik Schaefer, PhD, President, CEO & CSO, AssayQuant Technologies, Inc.

AssayQuant® has combined chelation-enhanced fluorescence with high-throughput peptide synthesis to create optimized physiological substrate sensors to monitor the activity of protein kinases and phosphatases. The result is a simple add & read method for continuous, quantitative and direct detection of activity using recombinant enzymes or crude lysates. A modified approach allows continuous monitoring of lipid kinases (DGKs). These enabling tools and services provide a quantum improvement to accelerate nextgen drug development.

1:05 pm Refreshment Break in the Exhibit Hall with Poster Viewing

CHASING DIFFICULT ONCOLOGY TARGETS

Terence H. Rabbitts, FRS, FMedSci, Professor, Molecular Immunology, Center for Cancer Drug Discovery, Institute of Cancer Research

Biologics (like intracellular antibodies) can be armed with warheads for targeted protein degradation (TPD) by fusing with E3 ligases. This is a flexible application of antibody engineering to target proteins in any location within the cell. Evidence will be presented of efficiency and effects of biologic TPD against specific RAS isoforms and the transcription complex formed by the chromosomal translocation protein LMO2. Biologics for TPD can be agnostic for binding site on the target protein.

2:10 pm

Targeting the Oncogenic Function of MYC with Bi-Functional Degrader Molecules

Elmar Wolf, Head, Biochemistry & Molecular Biology, University of Wuerzburg

The oncogene MYC presents a central unresolved challenge to drug discovery efforts. Being a transcription factor, MYC lacks catalytic activity and instead contains disordered regions that mediate macromolecular interactions. These features make it hard to develop compounds against MYC but offer the opportunity to inhibit MYC function by targeted degradation of MYC binding partners. We systematically identified MYC interacting proteins, analyzed their potential as cancer targets and developed bifunctional degraders.

Karteek Kadimisetty, Director, R&D, LifeSensors Inc.

PROteolytic TArgeting Chimeras (PROTACs) have emerged as a new class of protein degrader drug that can target the “undruggable” proteome for novel therapeutics. Currently, PROTAC discovery is hampered by large gap in understanding link between ubiquitination and degradation, and current methods to quantify this process are of limited utility in drug discovery. We addressed these gaps utilizing Tandem Ubiquitin Binding Entities (TUBEs) in high throughput biochemical and cell based assays.

Jessica Rich, Director, Commercial Development, Synthego

We will discuss how scalable genome engineering platforms can empower Target ID to Lead Optimization studies for TPD. This will include the time- and cost-effective creation of disease-relevant cell lines to validate targets with inducible protein loss and for easy assessment of protein turnover.

3:10 pm Refreshment Break in the Exhibit Hall with Poster Viewing
3:40 pm

Targeted Degradation of Transcription Factors by Transcription Factor Targeting Chimeras

Kusal Samarasinghe, PhD, Postdoctoral Associate, Department of Molecular, Cellular & Developmental Biology, Yale University

Many diseases, including cancer, stem from aberrant activation or overexpression of oncoproteins that are associated with multiple signaling pathways. Although proteins with catalytic activity can be successfully drugged, the majority of other protein families, such as transcription factors, remain intractable due to their lack of ligandable sites. Here, we report the development of TRAnscription Factor TArgeting Chimeras (TRAFTACs) as a generalizable strategy for targeted transcription-factor degradation. TRAFTACs are chimeric oligonucleotides that can simultaneously bind to the transcription factor-of-interest and to E3 ligase resulting in an induced degradation of the former via the proteasomal pathway.

4:10 pm

Targeted Protein Degradation – Where Are We Going?

Adam Gilbert, PhD, Senior Director, Discovery Sciences, Design & Synthesis Sciences, Pfizer Inc.

Overview of current state of targeted protein degradation and thoughts on where the field is heading with respect to clinical candidates, new degradation targets and different degradation strategies.

4:40 pm 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: Targeted Protein Degradation- Where Are We Going? 

Adam Gilbert, PhD, Senior Director, Discovery Sciences, Design & Synthesis Sciences, Pfizer Inc.
Matthew Calabrese, PhD, Senior Director & Head, Structural & Molecular Sciences, Pfizer Global R&D Groton Labs
5:25 pm Welcome Reception in the Exhibit Hall with Poster Viewing
6:25 pm Close of Day

Wednesday, September 29

7:30 am Registration Open and Morning Coffee

DEGRADING NOVEL TARGETS

7:55 am

Chairperson's Remarks

Corey Anderson, PhD, Director, Chemistry, C4 Therapeutics, Inc.
8:00 am

Targeted Protein Degradation of NTRK

Corey Anderson, PhD, Director, Chemistry, C4 Therapeutics, Inc.

Constituent activation of the tropomyosin receptor kinases, through aberrant fusions, leads to the unrestrained signaling of the oncogenic RAS/RAF and PI3K/AKT kinase cascade.  NTRK mutations or fusions have been identified as the driver of many cancers making the NTRK family of proteins an attractive target for drug discovery.  Herein we will describe the discovery and optimization of NTRK degraders.

8:30 am

Scalable Discovery and Streamlined Characterization of Novel Molecular Glue Degraders

Matthias Brand, PhD, Co-Founder and Vice President Biology, Proxygen GmBH

Molecular glue degraders are a novel therapeutic modality and induce destabilization of proteins that would otherwise be considered undruggable due to the lack of a ligandable pocket. Their discovery however has so far been serendipitous. The Proxygen glue degrader discovery platform enables rational screening for such molecules on an industrial scale. Here, we present a case study of the subsequent chemical optimization of the resulting hit molecules.

Vad Lazari, Director, Integrated Biology, Charles River
Will Esmieu, PhD

We developed HDAC Class IIa-potent and selective small molecule inhibitors suitable for testing in HD animal models. However, these inhibitors failed to replicate the beneficial effects observed in the genetic HDAC4 heterozygous loss of function phenotype in HD mouse models. As an alternative approach to the catalytic inhibition of HDAC4, we conjugated our best HDAC4 inhibitors from two different chemical series to a VHL (Von Hippel Lindau) E3 Ubiquitin Ligase ligand to generate PROTAC molecules which can be used to reduce total HDAC4 protein levels. Initially PROTAC molecules, from 2 chemical series differentiated by linker lengths and VHL binder attachment points, were screened for their ability to reduce HDAC4 protein levels in different cell types including mouse primary neurons. All compounds were potent degraders of HDAC4 protein. 

The best performing molecule was profiled in selectivity screening against other class IIa & class I HDACs and surprisingly, unlike the parent molecule, the HDAC4 PROTAC demonstrated exquisite selectivity for HDAC4 degradation vs other class IIa HDACs. We also present initial mouse in vivo data following ICV administration to determine whether sufficient distribution and target coverage could be obtained by this approach.

9:30 am Coffee Break in the Exhibit Hall with Poster Viewing
10:10 am

An IMiD-Inducible Degron Provides Reversible Regulation for Chimeric Antigen Receptor Expression and Activity

Seth Carbonneau, Principal Scientist II, Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research, Inc.

The recent development of successful CAR (chimeric antigen receptor) T cell therapies has been accompanied by a need to better control potentially fatal toxicities that can arise from adverse immune reactions. Here we present a ligand-controlled CAR system, based on the IKZF3 ZF2 b-hairpin IMiD-inducible degron, which allows for the reversible control of expression levels of type I membrane proteins, including CARs. Testing this system in an established mouse xenotransplantation model for acute lymphoblastic leukemia, we validate the ability of the CAR19-degron to target and kill CD19-positive tumor cells displaying complete control/clearance of the tumor. We also demonstrate that the activity of CAR19-degron can be regulated in vivo when dosing a FDA-approved drug, lenalidomide.

Jonathan Hall, PhD, Professor, Department of Chemistry and Applied Biosciences, Institute for Pharmaceutical Sciences, ETH Zurich

Proteolysis targeting chimeras (PROTACs) are heterobifunctional molecules composed of two domains. One domain binds selectively to a target protein of interest, the other domain sequesters elements of the ubiquitination machinery leading to intracellular proteolysis of the target. We are developing RNA PROTACs in which the recognition domain comprises the RNA binding element of an RBP in order to address this undruggable protein family.

11:10 am Transition to Plenary Keynote

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.

1:15 pm Enjoy Lunch on Your Own
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
1:55 pm Refreshment Break in the Exhibit Hall with Poster Viewing
2:35 pm Close of PROTACs and Targeted Protein Degradation – Part 1 Conference