Protein-Protein Interactions Icon

Cambridge Healthtech Institute’s Inaugural

Protein-Protein Interactions

Small Molecule Discovery for Difficult Drug Targets

4 - 5 DECEMBER 2024

 

 

Protein-protein interactions (PPIs) as drug targets are disease-relevant complexes of proteins bound to one another, and sometimes to DNA or RNA as well. PPIs are difficult for medicinal chemists to design or discover drugs against because there is no enzymatic site to inhibit. Furthermore, the PPIs usually have flat interaction surfaces making it harder for small molecules to bind the PPIs. At CHI's Protein-Protein Interactions conference join a global community of medicinal, biophysical and structural chemists to share insights, successes and lessons learned from working on these hard-to-drug targets.

Wednesday, 4 December

PLENARY KEYNOTE SESSION

11:00

Welcome Remarks

Anjani Shah, PhD, Senior Conference Director, Cambridge Healthtech Institute

11:15

PLENARY KEYNOTE: How Protein Degraders Work: Molecular Mechanism and Design Principles 

Alessio Ciulli, PhD, Professor, Chemical & Structural Biology and Director of the Centre for Targeted Protein Degradation, University of Dundee

Our laboratory uses molecular information on protein-protein interactions and protein degradation to discover novel therapeutics. Degrader molecules, also known as PROTACs (PROteolysis-Targeting Chimeras) recruit proteins to E3 ligases for targeted protein degradation. Formation of a ternary complex—amongst the PROTAC, the E3 and the target—leads to the tagging of the target protein by ubiquitination, and subsequent proteasomal degradation. This fundamental understanding has enabled us to develop further small molecules for hard-to-target proteins and shown how to improve PROTAC activity.

12:00Networking Lunch in the Exhibit Hall

TARGETING RAS & INTERACTING PARTNERS

13:15

Chairperson's Remarks

Adrian L. Gill, PhD, Senior Vice President, Medicinal Chemistry, Revolution Medicines

13:20

Identification and Optimisation of a Novel Series of KRAS G12C Inhibitors Leading to the Discovery of JDQ443 and Beyond

Simona Cotesta, PhD, Executive Director Medicinal Chemistry, Novartis Biomedical Research

RAS is the most frequently mutated oncogene in cancer, with KRAS G12C mutations most commonly found in lung adenocarcinoma and colorectal cancer. Covalent inhibitors of KRASG12C have shown antitumour activity against advanced/metastatic KRAS G12C-mutated cancers. Here we report the identification of a new class of pyrazole-based KRASG12C inhibitors discovered by structure-based de novo design, the hit-to-lead, and lead optimisation leading to the discovery of JDQ443 and beyond.

13:50

Discovery of a Novel Class of Brain-Penetrant Inhibitors of KRASG12C

Doyle Cassar, PhD, Senior Research Scientist, Medicinal Chemistry, AstraZeneca

The glycine to cysteine mutation at codon 12 of KRAS represents an Achilles' heel that has now rendered this important GTPase druggable. Herein we report our structure-based drug design approach that led to the identification of AZD4747, a clinical development candidate for the treatment of KRASG12C-positive tumours, including treatment of CNS metastases. AZD4747 is a highly potent and selective inhibitor of KRASG12C with an anticipated low clearance and high oral bioavailability profile in humans.

14:20Poster Spotlight: Oral Presentation of Selected Poster(s) In-Person Only

14:50Refreshment Break in the Exhibit Hall with Poster Viewing

15:30

FEATURED PRESENTATION: Tackling KRAS with Complementary Modalities—Discovery of PROTACs and Small Molecule Inhibitors

Tobias Wunberg, PhD, Director Medicinal Chemistry, Medicinal Chemistry, Boehringer Ingelheim RCV GmbH & Co. KG

Activating mutations in KRAS-G12 are common in many cancers, including lung, colon, pancreas, and other tumours and is one of the most frequent drivers of human malignancies. These activating mutations lead to an increase in GTP-KRAS, allowing binding to effector proteins and triggering of downstream signaling pathways which regulate cell proliferation, differentiation, survival, and tumour progression. We will describe the discovery of our recently published GDP-KRAS inhibitors BI-2865 and BI-2493 (Kim et al., Nature, 2023) and KRAS PROTAC ACBI3 (Popow et al., bioRxiv, 2023). Based on in vivo results, we will also discuss a head-to-head comparison of these two modalities.

16:00

Discovery of RMC-9805, an Oral RAS(ON) G12D-Selective Covalent Tri-Complex Inhibitor

John E. Knox, PhD, Vice President, Structural Chemistry & Discovery Sciences, Revolution Medicines

The investigational agent RMC-9805 is an orally bioavailable, RAS(ON)G12D mutant-selective covalent inhibitor that forms a tri-complex between cyclophilin A and the “ON” state of RASG12D, enabling selective covalent engagement of Asp-12 and disruption of RAS signaling. RMC-9805 employs a warhead with low intrinsic reactivity and relies on a neomorphic protein interface to help catalyse the covalent reaction. RMC-9805 exhibits anti-tumour activity across a panel of human RASG12D cancer preclinical models.

16:30

Development of Orally Bioavailable RAS-Inhibitory Peptide Guided by Drug-Like Criteria

Atsushi Ohta, PhD, Head of Modality Technology Department, Chugai Pharmaceutical Co., Ltd.

Macrocyclic peptides have been attracting attention as a scaffold of protein-protein interaction inhibitors; however, limited success has been reported as an oral drug. Here, we report a methodology for creating a cell-permeable and orally bioavailable peptide drug by identifying important factors for better drug-likeness and developing library technologies affording highly N-alkylated cyclic peptides. As an example, the discovery of a RAS inhibitory clinical compound (LUNA18) will be presented.

17:00Close of Day

Thursday, 5 December

08:00Registration and Morning Coffee

MOLECULAR GLUES BEYOND DEGRADERS

08:25

Chairperson's Remarks

Marianne Schimpl, PhD, Associate Principal Scientist, Structural Biology, AstraZeneca

08:30

Non-Degrading Molecular Glue Platforms for Targeting Hard-to-Drug Proteins

Rick Ewing, PhD, Vice President and Head of Chemistry, Rapafusyn Pharmaceuticals

Type I molecular glues from nature, FK506, Cyclosporin, and Rapamycin, have resulted in marketed therapies. The complex structures of these molecules have limited application to only a few protein targets. The presentation will describe the construction of large DEL and array libraries of type I molecular glues used for finding chemical starting points for a wide range of intracellular targets. Concepts of topological diversity, of optimization studies for cell permeability and of molecular glue characterization will be described.

09:00

Computational Approaches to Finding PPIs as Starting Points for Molecular Glue Projects

Sergio Martinez Cuesta, PhD, Associate Director in Bioinformatics, AstraZeneca

We are building a bioinformatics and data integration platform to enable the discovery of effectors for molecular glue projects (e.g., E3 ligases for undruggable targets). This talk will offer an overview of our internal capabilities and systems, as well as external collaborations. It will feature an example to showcase the practical application of these elements, offering insights into our approach.

09:30In-Person Breakout Discussion Groups

In-Person Breakouts are informal, moderated discussions, allowing participants to exchange ideas or experiences, develop collaborations around a focused topic, and meet scientists with similar interests. Each breakout will be led by facilitators who keep the discussion on track and the group engaged. Breakout discussion topics and moderators will be listed soon.

IN-PERSON ONLY BREAKOUT 4:

Macrocycle Drug Discovery

Maxwell D Cummings, PhD, President, The Dinosaur Project, LLC

  • Peptides and synthetic small-molecules
  • Macrocyclization: before, during or after hit discovery?
  • Computational approaches to understanding bRo5
IN-PERSON ONLY BREAKOUT 5:

Applying Fragment-Based Drug Design to PPI Drug Discovery

Engi Hassaan, PhD, Associate Editor, Nature Communications

  • Innovation in fragment screening approaches: can we accurately screen fragments in cells; can we screen from crude compound mixtures? 
  • The best methods to validate fragment binding and measure fragment affinity
  • The promise of AI: what else can it do? Discover hot spots, predict fragment binding affinities, filter out false positives, pre-screen large libraries, design leads from fragments?
IN-PERSON ONLY BREAKOUT 6::

Covalent Compounds in Drug Discovery

Giulia Alboreggia, PhD., Postdoctoral Fellow, Pellecchia Lab, University Of California Riverside

  • Which amino acid to target beyond the cysteine and which electrophile to use
  • Ligand-first (peptide mimetic or small molecule) vs electrophile-first approach​
  • Techniques for covalent screening and validation

10:15Networking Coffee Break

NEURODEGENERATION & VIRAL PPI TARGETS

10:45

γ-Secretase Modulators (GSMs)

Rosa Maria Rodriguez Sarmiento, PhD, Expert Scientist & Project Lead, Medicinal Chemistry, Roche Pharma

Our initial efforts to obtain a novel and well-balanced γ-secretase modulator with the potential to become an oral disease-modifier for early AD will be presented.

11:15

Druggable Pockets in β-Glucocerebrosidase (GCase), a Parkinson's Disease Drug Target

Christopher Agnew, PhD, Project Lead, Astex Pharmaceuticals Ltd.

Mutations in the lysosomal protein GCase have been linked to increased risk for Parkinson’s Disease. Therapeutics aimed at enhancing GCase protein stability, trafficking, and activity are therefore of high relevance. Fragment screening by X-ray crystallography and other biophysical methods enabled the identification of new potentially druggable sites on the GCase protein surface. Compound elaboration produced high-affinity molecules able to positively modulate GCase activity in cells.

11:45

Pursuing Targeted Protein Degradation for Investigating CNS Diseases

Nur Kocaturk, PhD, Post Doctoral Fellow, Center for Targeted Protein Degradation, University of Dundee

Diseases of the central nervous system, including neurodegenerative, neurological, and cancer-related diseases account for hundreds of millions of patients worldwide with poor or no treatment options. Furthermore, the validation of new therapeutic concepts in these areas suffers from a lack of potent and selective chemical tools. This talk will introduce work the Farnaby Lab is conducting, within the Dundee Centre for Targeted Protein Degradation, to understand how we can increase our ability to identify CNS active small molecule degraders.

12:15

Antiviral PPI Inhibitors: Invention of the HIV-1 Maturation Inhibitor VH3739937 (VH-937)

Alicia Regueiro-Ren, PhD, Scientific Senior Director, Medicinal Chemistry, Bristol Myers Squibb Co.

I present discovery of VH3739937, an HIV-1 maturation inhibitor (MI) in clinical trials. HIV-1 maturation inhibitors (MIs) bind to the viral Gag polyprotein, preventing the final HIV-1 protease-mediated cleavage of Gag from occurring. VH3739937 is an MI with nanomolar activity vs. viruses with substitutions and deletions, conferring a much reduced sensitivity to previous MIs. In a Phase I study in healthy volunteers, VH-937 was found to be safe and demonstrated pharmacokinetic properties supporting QW dosing in an ongoing Phase II study in infected subjects.

12:45Networking Luncheon

bRo5 MOLECULES FOR PPIs: PERMEABLE MACROCYCLICS & ORAL PEPTIDES

13:45

Chairperson's Remarks

Armon Sharei, PhD, Founder & CEO, Portal Biotechnologies

13:50

Macrocyclic Target Binding and Cell Permeability in bRo5 Space

Jan Kihlberg, PhD, Professor, Organic Chemistry, Uppsala University

The ability of macrocycles to adopt disc- and sphere-like conformations allows them to modulate difficult-to-drug targets. Consequently, macrocyclic drugs frequently reside in the beyond-rule-of-5 space where solubility, cell permeability, metabolic stability, and thereby oral bioavailability is challenging. Physics-based models provide mechanistic insight into cell permeability, (e.g., into molecular chameleonicity), while models developed by machine learning allow accurate differentiation of macrocycles that have high or low cell permeability.

14:20

FEATURED PRESENTATION: Discovery and Development of MK-0616: Macrocyclic Oral PCSK9 Inhibitor

Abbas M. Walji, PhD, Senior Director, Discovery Chemistry Modalities, Merck & Co., Inc.

PCSK9 is a secreted serine protease implicated in the progression of hypercholesterolemia. Inhibitors of the PCSK9: LDL receptor have demonstrated clinical benefit in the reduction of LDL cholesterol and are currently used in the treatment of dyslipidemia. However, the current approved therapies which include two monoclonal antibodies and one siRNA, are limited due to administration by injection. This presentation will highlight Merck’s discovery program culminating in the discovery of MK-0616, and the clinical translation of oral PK/TE/PD in Phase 1 SAD/MAD studies.

14:50

Macrocyclic Peptides: Passively Permeable, Potent, and Oral Inhibitors of PPIs

Cameron Pye, PhD, CEO and Co-Founder, Unnatural Products

Macrocyclic peptides can achieve passive permeability and oral bioavailability on par with small molecules. However, unlike small molecules, they are able to bind shallow, protein-protein interaction (PPI) interfaces with exquisite potency and and selectivity, opening up a new swath of targets and mechanisms-of-action inside and outside the cell. This presentation will showcase examples of successful intracellular PPI inhibitors derived from the Unnatural Products platform.

15:20Close of Conference






Save the Date
Next-Gen Degraders & Molecular Glues
Protein-Protein Interactions

Learn About Our U.S. Event
Register Early and Save