Protein-Protein Interactions Icon

Cambridge Healthtech Institute’s 17th Annual

Protein-Protein Interactions

Small Molecule Lead Discovery and Optimization for Difficult Drug Targets

APRIL 3 - 4, 2024

 

Cambridge Healthtech Institute's Protein-Protein Interactions (PPIs) conference covers discovery or design, and optimization of compounds that either disrupt or stabilize protein-protein or protein-nucleic acid complexes that cause disease. These complexes are also often intracellular and therefore only amenable to cell-penetrating compounds such as small molecules and newer modalities like (macro)cyclic peptides. However, they have been typically considered ‘difficult-to-drug' because their flat interaction surfaces make the PPIs harder for small molecules to gain a foothold. Furthermore, they are not typically enzymes, so traditional high-throughput enzymatic assay-based screening for identifying small molecule drug leads hasn't been possible. Recent advances that have spurred progress include: biophysical techniques that screen for molecular interactions rather than an enzymatic event, covalent drug discovery strategies, fragment-based ligand approaches, and structure-based drug design. Induced proximity and targeted protein degradation (TPD) 'destroy what you can't inhibit' approaches have also furthered the field and will be covered tangentially. Join fellow medicinal, biophysical, and structural chemists to learn the latest, share insights, and discuss strategies.

Wednesday, April 3

Registration Open12:00 pm

Welcome Remarks1:30 pm

NON-DEGRADER GLUES, (DE)-STABILIZERS, AND TARGETING RAS

1:35 pm

Chairperson's Remarks

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

1:40 pm

Multi-Ras Inhibitors

Anne Edwards, PhD, Senior Scientist I, Revolution Medicines

RAS oncogenes are among the most frequently mutated genes in cancer, with common driver mutations occurring at codons 12, 13, and 61. Here, we describe RMC-7977, a potent, oral, non-covalent, tri-complex RASMULTI(ON) small molecule inhibitor with broad spectrum activity for the active state of both mutant and wild-type (WT) KRAS, NRAS, and HRAS variants (a RASMULTI(ON) inhibitor). Within cells, RMC-7977 forms a tri-complex along with RAS(ON) and cyclophilin A (CypA), driving a near-immediate disruption of RAS effector binding and extinction of RAS(ON) signaling through steric occlusion. 

2:10 pm

Discovery of MRTX1719: A Synthetic Lethal Inhibitor of the PRMT5/MTA Complex for the Treatment of MTAP-Deleted Cancers

Chris Smith, PhD, Executive Director, Drug Discovery, Mirati Therapeutics

The methylthioadenosine phosphorylase (MTAP)-encoding gene is co-deleted with p16/CDKN2a in ~10-15% of all human cancers, leading to elevated levels of the MTAP substrate methylthioadenosine (MTA) in these cancers. MTA binds Protein Arginine N-Methyl Transferase (PRMT5) to form the PRMT5/MTA complex. We describe biophysics-aided discovery of a 4-(aminomethyl)phthalazin-1(2H)-one fragment and its evolution into MRTX1719, a clinical-stage, selective inhibitor of the PRMT5/MTA complex as a potential precision medicine for treating MTAP-deleted tumors.

2:40 pm A Live Cell PRMT5 NanoBRET™ Target Engagement Assay to Quantify Competitive and Uncompetitive Modes of Inhibition

James Vasta, PhD, Senior Research Scientist, Research & Development, Promega Corporation

PRMT5 is an essential arginine methyltransferase that uses SAM as the methyl donor. Here we describe a novel NanoBRET™ Target Engagement assay that enables characterization of PRMT5 inhibitors with diverse inhibitory mechanisms in living cells. Both substrate- and SAM-competitive engagement are quantifiable. Moreover, MTA-uncompetitive engagement can also be quantified, facilitating the development of inhibitors that exploit the accumulation of MTA in various cancers by binding cooperatively to the PRMT5/MTA complex.

Refreshment & Dessert Break in the Exhibit Hall with Poster Viewing3:10 pm

4:00 pm

Measuring Stable Interactions by SPR: Addressing the Long Residence-Time Challenge

Thomas P. Garner, PhD, Principal Scientist, Biophysics, Genentech, Inc.

Many biomolecular interactions form very long-lived complexes. These can be extremely challenging to measure leading to unfavorably long incubation times in biochemical/biophysical assays and inaccurate measurements. SPR has the advantage of measuring kinetics with high accuracy, the “chaser” method extends the range of measurable half-lives with high accuracy but are low throughput. We present adjustments to the chaser assay to improve throughput and its application to measuring protein interactions.

4:30 pm

Targeted Degradation by Protein Destabilizing Compounds

Maurizio Pellecchia, PhD, Professor, Biomedical Sciences Division, University of California, Riverside

Targeted protein degradation is based on bifunctional ligands to recruit the ubiquitin-proteasome system. As an alternative strategy, we explored here the idea to design protein degraders based on the section of ligands that cause protein destabilization, and that in turn induce target degradation in cell. In an application of this approach, we found that agents can act as molecular crowbars that, destabilizing critical intramolecular interactions, cause protein degradation in cell.

In-Person Breakouts5:00 pm

In-Person Breakouts are informal, moderated discussions, allowing participants to exchange ideas and experiences and develop future collaborations around a focused topic. Each breakout 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 Breakout Discussions page on the conference website for a complete listing of topics and descriptions.

IN-PERSON BREAKOUT 4:

Covalent Drug Discovery (SESSION ROOM)

Nir London, PhD, Senior Scientist, Organic Chemistry, Weizmann Institute of Science

Maurizio Pellecchia, PhD, Professor, Biomedical Sciences Division, University of California, Riverside

  • Starting points for covalent drug discovery—potent reversible binders vs. covalent fragment hit
  • sStrategies to target nucleophilic amino acids beyond cysteine
  • Characterization and prioritization of novel warheads​​​
IN-PERSON BREAKOUT 5:

Emerging Technologies for Addressing PPIs (FOYER)

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

  • Discovery of stabilizers or non-degrading molecular glues 
  • Targeting PPIs with macrocyclic peptides: advantages and challenges
  • Technologies for discovering PROTACs and molecular glue degraders​
IN-PERSON BREAKOUT 6:

Biophysical Tools for Targeting PPIs (FOYER)

Elisa Barile, PhD, Principal Scientist, Structural Biology & Biophysics, Takeda, San Diego

Phillip Schwartz, PhD, Director, Biophysics, Septerna

  • Best orthogonal biophysical and biochemical approaches: XRC, Cryo-EM, NMR, SPR/GCI, DSF, FRET, MS, FP, and more 
  • Application of biophysics in alternative modalities such as Protac, glues, RNA modulators
  • Testing funnels: which techniques to use, and where? (e.g., assessing lead potency vs. tracking PPI affinity shifts...)​

Close of Day5:45 pm

Dinner Short Course Registration5:45 pm

Dinner Short Course*6:15 pm

SC6: Principles of Drug Design: Ligand-Receptor Interactions and More

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

Thursday, April 4

Registration Open7:15 am

Diversity in Chemistry Breakfast Discussion7:45 am

Grab a plate and then a seat to join one of the in-person discussions below on growing the enterprise of chemistry (in terms of people diversity, not molecules). This session originated 4 years ago with a focus on ‘Women in Chemistry’, but every year the discussions raised more issues than time allowed. We’re broadening the topics but breaking them into smaller discussion-focused groups; topics will include the below. Please visit the Breakout Discussions page on the conference website for more details.


Paternity and Extended Leave  Moderator(s): Thomas Garner, Genentech 

Advancing Women in Chemistry  Moderator(s): Katerina Leftheris, Vilya

Diversity, Equity, and Inclusion Efforts at Institutions & Companies  Moderator(s): Michelle Arkin, UCSF


PLENARY KEYNOTE SESSION

8:30 am

Plenary Welcome Remarks from Lead Content Director with Poster Finalists Announced

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

8:35 am

PLENARY KEYNOTE: Reimagining Druggability Using Chemoproteomic Platforms

Daniel Nomura, PhD, Professor of Chemical Biology and Molecular Therapeutics, Department of Chemistry, University of California, Berkeley

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 applying chemoproteomic platforms to discover and pharmacologically target unique and novel ligandable hotspots for disease therapy.

Coffee Break in the Exhibit Hall with Poster Viewing and Best of Show Awards Announced9:20 am

PPI-TARGETING TOOLS AND INNOVATIONS

10:10 am

Chairperson's Remarks

Heike Wobst, PhD, Senior Scientist, Jnana Therapeutics

10:15 am

Targeted Autophagy for Degradation of Aberrant PPI Complexes

Chang Hoon Ji, PhD, Executive Director, Bio R&D Center, AUTOTAC Bio, Inc.

Protein complexes and many non-protein targets are degraded in the lysosome via autophagy. This is in contrast to the ubiquitin proteosome system which is the 'final' destination of proteins. I describe our targeted protein degradation platform for lysosome: the AUTOphagy-TArgeting Chimera (AUTOTAC) TPD platform was used to selectively degrade pathological aggregates, and combat neurodegeneration-associated pathophysiology.

10:45 am

Affinity Selection-Mass Spectrometry (ASMS) Applicability for PPI Targets

Hans-Peter N. Biemann, PhD, Distinguished Scientist, Integrated Drug Discovery, Sanofi

Affinity Selection-Mass Spectrometry (ASMS) identifies novel small molecule ligands for soluble and membrane proteins via a mass-encoded readout. We have recently applied ASMS to several challenging targets across different protein classes. These target proteins generally lack recognized druggable clefts and include PPI moieties. This presentation will review the process and status of hit ID comprising distinct externalized library HTS as well as reaching into of Sanofi’s internal collection via an intermediate virtual screening step.

11:15 amPoster Spotlight:

P035: Analysis of HDAC6:Tubulin Interaction Using Affinity Selection Mass Spectrometry
Ryan Kilburn, Eli Lilly and Company

11:30 am

Proteomic Discovery of Chemical Probes that Modulate Protein Complexes in Human Cells

Jarrett R. Remsberg, PhD, formerly of Cravatt Lab, Scripps Research Institute; Scientist, Proteomics, Belharra Therapeutics

Most human proteins lack chemical probes, and while several large-scale and generalizable small-molecule binding assays have been introduced, how compounds discovered in "binding-first" assays affect protein function often remains unclear. Here, we describe a "function-first" proteomic strategy that uses size exclusion chromatography (SEC) in conjunction with cysteine-directed activity-based protein profiling to identify changes in protein-protein interactions, including stereoselective engagement of SF3B1, stabilizing a dynamic state of the spliceosome.

12:00 pm

Inducing Conformational Change: Could AI Have Discovered this PPI Inhibitor?

Jonathan B. Baell, PhD, Executive Director, Early Leads Chemistry, Lyterian Therapeutics

Peptidomimetic design to mimic protein-protein interaction hotspot is a logical approach to PPI inhibitor discovery. We had done so for the BCL-XL -BH3 binding groove by mimicking important BH3 protein binding residues. Unexpectedly, more optimized ligands induced an entirely unexpected conformation of BCL-XL. In light of the current AI debate, we revisit this observation as a challenging benchmark to which AI-enabled PPI discovery should aspire.

Enjoy Lunch on Your Own12:30 pm

Refreshment Break in the Exhibit Hall with Poster Awards Announced1:05 pm

ALLOSTERIC MODULATORS

1:55 pm

Chairperson's Remarks

Robert D. Mazzola, PhD, Director & Principal Scientist, Chemical Research, Merck & Co.

2:00 pm

Fragment-Based Discovery of Allosteric SHP2 Inhibitors

Tom Davies, PhD, Director, Molecular Sciences, Astex Pharmaceuticals

We present the application of FBDD to the oncology target SHP2 phosphatase. Hits at an allosteric site were evolved using structure-based design to a low-nanomolar lead which inhibits tumour growth in xenografts. Phosphatases have been deemed undruggable due to their polar and conserved active sites, and we highlight the ability of fragment-based screening to detect hits in novel pockets which can be exploited to identify differentiated modulators for challenging targets.

2:30 pm

Targeting CBM, a Tri-Protein Signaling Hub, by Inhibition of MALT1 for the Treatment of B Cell Lymphomas

Susanta Samajdar, PhD, CSO, Aurigene Discovery Technologies Ltd.

MALT1 is a key regulator of antigen-receptor signaling, wherein it partners with BCL10 and CARMA1 to form the CBM complex in which protease activity of MALT1 cleaves the negative regulators leading to activation of NF-kB. Constitutive activation of NF-kB is a key driver in B cell lymphomas. We have identified a development candidate that inhibits MALT1 with the “best-in-class” profile including potent activity in whole blood and selectivity over off-targets.

3:00 pm PLENARY PANEL DISCUSSION:

Innovative Drug Discovery: Insights from Venture Capitalists

PANEL MODERATORS:

Michelle Arkin, PhD, Chair and Distinguished Professor, Pharmaceutical Chemistry & Director, Small Molecule Discovery Center, University of California, San Francisco

Daniel A. Erlanson, PhD, Chief Innovation Officer, Innovation and Discovery, Frontier Medicines Corporation

The high-risk but 'high impact-when-successful' strategy of VC investors gives them a uniquely critical lens through which to view innovation. Join us for an interactive discussion with VCs who will share the trends they are watching in drug discovery. The panel represents a variety of small and large venture firms, who provide early rounds of funding, as well as those who invest at later or all stages.

Topics to be covered:
  • Introduction to VC panelists and their fund’s areas of focus
  • Investing in platforms versus products
  • Perspectives on emerging technologies or approaches (AI/ML, induced proximity and more)
  • Advice on funding options for start-ups beyond VCs, such as angels and grants
  • Pitfalls for early-stage companies to avoid when seeking funding
PANELISTS:

Wendy B. Young, PhD, BioPharma Discovery

Rebecca Silberman, PhD, Senior Venture Associate, RA Capital Management LLC

Shyam Masrani, Principal, Medicxi

Jamie Kasuboski, PhD, Partner, Luma Group

Olga Danilchanka, PhD, Principal, MRL Ventures Fund

Networking Refreshment Break3:45 pm

TARGETED COVALENT INHIBITORS

4:00 pm

Ligand Efficiency Metrics in Covalent Drug Discovery

Benjamin Horning, PhD, Scientist, Vividion Therapeutics

Ligand efficiency metrics have made a major impact in medicinal chemistry, from the prioritization of early hits through lead optimization. With the ascendancy of covalent inhibitors in drug discovery, it is important to consider how to translate efficiency metrics to covalent modalities. Herein, we introduce ligand reactivity efficiency (LRE) as a means of correcting potency for intrinsic electrophilic reactivity, and explore kinetic regimes in which this analysis is appropriate.

4:30 pm

Small Molecule-Targeted Covalent Inhibitors of the HEG1-KRIT1 Protein-Protein Interaction

Carlo Ballatore, PhD, Professor, Pharmaceutical Science, University of California San Diego

The protein-protein interaction (PPI) between HEG1 (Heart of glass 1) and KRIT1 (Krev interaction trapped 1) plays an important role in controlling vascular development and permeability. Small-molecule modulators of this PPI may be useful as candidate therapeutics and/or pharmacological tools. Here we report the identification/characterization of hydroxy-naphthaldehyde (HNA) fragments that act as targeted covalent reversible ligands of a noncatalytic Lys of KRIT1 with high specificity and long residence time (>8h) resulting in inhibition of the PPI in cell-free and cell-based assays.

5:00 pm

FEATURED PRESENTATION: Lysine-Targeted Covalent Protein Reagents

Nir London, PhD, Senior Scientist, Organic Chemistry, Weizmann Institute of Science

Installing a covalent electrophile on a peptide or protein-based scaffold with an extended binding footprint enables the targeting of shallow protein surfaces not typically addressable using small molecules. We report protein-based thio-methacrylate esters: electrophiles with a diverse reactivity profile that can be installed easily on unprotected peptides and proteins via cysteine side chains, and react efficiently and selectively with cysteine and lysine side chains on the target.

Close of Conference5:30 pm