Skip to main content

Partnering

Partnering Opportunities

The focus of our research is on drug discovery using fragment-based methods and structure-based

design. (Fesik Lab homepage).

 

Dr. Fesik pioneered the use of fragment-based methods for discovering high affinity ligands

for proteins over 25 years ago (1) and has applied this method to several protein targets.

After 26 years in drug discovery at Abbott/Abbvie with the last nine-years serving as

Divisional Vice President of Cancer Research, Dr. Fesik was recruited by Vanderbilt

University in 2009 to be a professor in Biochemistry (primary appointment), Pharmacology,

and Chemistry. In addition, many of the Fesik lab members have also worked in the

Pharmaceutical Industry, and we apply the same principles, approaches, thought processes,

and techniques used in drug companies with an emphasis on targeting “undruggable”

targets. As shown in the project descriptions, we have been quite successful over the last

several years that has led to projects at various stages ranging from hit identification to

clinical candidates. Although KRAS is partnered with Boehringer Ingelheim, the other

projects described below are available for partnering.

 

 If you are interested in working with us on any of the projects or are interested in hearing

more about these and other ongoing projects in the lab, don’t hesitate to contact Stephen

Fesik via email at stephen.fesik@vanderbilt.edu.

 

Cancer


KRAS
(partnered with BI)

KRAS is a GTPase that is heavily mutated in many cancers, including 86-96% of pancreatic

cancers, 40-54% of colorectal cancers, and 27-39% of lung cancers. Activating mutations in

KRAS increase signaling in several important cellular pathways and are the most common

oncogenic drivers in human cancer. Thus, KRAS is a highly validated cancer target.

However, despite many attempts to target KRAS over decades, it was long thought

impossible to drug. Recent efforts, including the approval of G12C KRAS inhibitors, have

changed this way of thinking.

 

Our lab has a 15-year track record of targeting KRAS. Using NMR-based fragment screens,

we identified small molecules that bind to KRAS in the active GTP- and inactive GDP-bound

forms (2). Subsequent optimization of the fragment hits that bind to KRAS-GTP using

structure-based design led to a compound that binds to the switch I/II site with nanomolar

affinity, inhibits all GEF, GAP, and effector interactions with KRAS, and displays an

antiproliferative effect in KRAS mutant cells (3). We attempted to optimize fragments that

bind to KRAS-GDP by using a second site screen to identify compounds that could be linked

or merged to the first site hits. However, the molecules simply displaced the first site ligand

in the screen. We therefore developed a strategy to hold the first site ligand in place by

introducing cysteine mutants near the switch I/II site and identifying a molecule that could

covalently attach to these cysteines (4). Using S39C KRAS with a covalently bound switch

I/II blocker, we performed a second site screen and identified 20 hits that bound to KRAS at

a second site. We obtained X-ray crystal structures that showed these hits unexpectedly

bind to the switch II site. Optimization using structure-based design led to BI 1823911, a

G12C inhibitor in phase I (5), a pan KRAS inhibitor (BI 3706674) which has recently entered

phase I (6), a pan-RAS degrader (7), and mutant selective KRAS inhibitors that are currently

in preclinical studies. Some of this work was performed under a collaborative licensing

agreement with Boehringer Ingelheim. Interestingly, the reported KRAS inhibitors/degraders

under development from BI all contain the fragment that we identified from the second site

screen (Fig. 1).

 

MYC

MYC is a transcription factor that is overexpressed in most cancers. MYC is composed of

an N-terminal transactivation domain, a C-terminal basic-helix-loop-helix leucine zipper DNA

binding domain, and a central region (Fig. 2). It forms a heterodimer with MAX, binds to Ebox

DNA, and drives

the expression of

genes required for cell

growth, proliferation,

metabolism, genome

instability, and

apoptosis. Like KRAS,

MYC is a highly

validated target but is

thought to be difficult or

impossible to drug.

Unlike KRAS, however, no molecules that directly target MYC have been approved to date.

Our first attempt to drug MYC used an NMR-based fragment screen of the transactivation

domain and the DNA binding domain of MYC – two regions of the protein required for activity.

No confirmed hits were found for either of these proteins. This outcome was not surprising,

as MYC, in the absence of MAX, is intrinsically disordered in solution as evidenced by the

poor chemical shift dispersion observed in our NMR spectra. Thus, due to the lack of

confirmed hits, we turned to other approaches for targeting MYC.

 

MYC/WDR5 (available for partnering)

Using a two-hybrid and proteomic screen,

Professor Bill Tansey (Vanderbilt) discovered that

the central portion of MYC (MYC box IIIb) binds to

WDR5, and this interaction is required for MYCdriven

tumorigenesis (8). These data led us to our

second approach for targeting MYC using a

cofactor of MYC, WDR5, which is much more

druggable.

 

From a fragment-based screen of WDR5, we

identified hits that bound to the site where MYC

interacts with WDR5 (WBM site) (9) and hits that bind to the opposite end of the protein at

the site where MLL1 binds to WDR5 (WIN site) (10) (Fig. 3). From these fragments, we

discovered potent chemical probes at both the WBM (9,11) and the WIN sites (10,12) that

allowed us to study the biology of WDR5 and evaluate WDR5 as a cancer drug target. We

found that WDR5 facilitates the recruitment of MYC to chromatin to control the expression

of genes linked to ribosome biogenesis—a critical tumor-sustaining function of MYC.

Importantly, disrupting the MYC-WDR5 interaction promotes tumor regression in vivo,

validating the importance of WDR5 for tumor maintenance by MYC (13). WIN site inhibitors

act by displacing WDR5 from chromatin at ribosomal protein genes (14) and not by changes

in histone methylation as previously thought (15). These findings indicate that a WDR5

inhibitor may be useful for treating a wide range of cancers and encouraged us to further

optimize the chemical probes into drugs. Upon extensive optimization, we discovered single

digit picomolar WDR5 inhibitors that are orally bioavailable, efficacious in vivo, and safe (16-

18) (Fig. 4). We have selected a candidate for IND-enabling studies and are currently

working with the NCI to advance this molecule into the clinic.

 

MYC/MAX (available for partnering)

Inhibiting the binding of MYC to DNA represents another method of targeting MYC.

OmoMYC, a protein-based inhibitor of DNA-binding to MYC, has been shown to promote

tumor regression in a host of cancer types in vivo with only limited and reversible toxicities

(19). However, protein-based inhibitors typically face development challenges and often do

not make ideal drugs. Small, drug-like molecules that bind to the MYC/MAX dimer and

prevent it from recognizing its cognate DNA binding sites would disable all or most of the

transcriptional functions of MYC in cancer cells, promote tumor regression, and have

superior drug-like properties compared to a protein-based agent like OmoMYC. Therefore,

we set out to identify compounds that block the binding of MYC to DNA.

 

Unlike MYC alone, the MYC/MAX dimer adopts a folded structure and therefore may be

more druggable. The MYC/MAX dimer can be labeled and expressed in E. coli, and its 1H

and 15N NMR resonances have recently been assigned. Using an NMR-based fragment

screen of the MYC/MAX dimer, we identified multiple hits. X-ray crystal structures of two of

these hits when bound to the MYC/MAX dimer revealed that these hits bind at the interface

of the MYC/MAX dimer with DNA (Fig. 5). Furthermore, these molecules and their analogs

were found to weakly disrupt DNA binding by MYC (Fig. 5). We have now obtained multiple

X-ray structures of the MYC/MAX dimer when bound to additional hits and their analogs and

have used structure-based design to improve binding by 1000-fold. Current work involves

exploring the ability of these compounds to inhibit the functions of MYC using assays set up

by Bill Tansey and testing our inhibitors for treating MYC-driven cancers in vivo.

 

WNT pathway: b-catenin (available for partnering)

The Wnt pathway is a highly validated drug discovery target for colorectal cancer (CRC) and

other malignancies (20). Mutations in APC, AXIN, or b-catenin occur in 90-95 % of all CRCs,

leading to excessive cytosolic b-catenin levels. Dysregulation of Wnt signaling is recognized

as the initial, causative event in most CRC cases and has been shown to support tumor

growth. However, despite decades of research, direct inhibition of the Wnt pathway

downstream of activating mutations has been difficult to achieve. Our project aims to

discover potent and selective inhibitors of the Wnt pathway via targeted protein degradation

of b-catenin, using heterobifunctional molecules that bind directly to b-catenin.

We have used an NMR-based fragment screen to identify small molecules that bind bcatenin

and identified fragments that bind to a site not shared by its known endogenous

binding partners. More than 100 high-resolution crystal structures of analogs of these

fragments bound to b-catenin have been determined and used to inform structure-based

optimization of ligands for b-catenin. We have discovered compounds that bind tightly (KD

<20 nM) to b-catenin and contain solvent-facing functional groups suitable for use in

heterobifunctional Proteolysis Targeting Chimeras (PROTACs). Using these b-catenin

ligands, we have created PROTACs that recruit the E3 ligase CRBN into a ternary complex

with b-catenin, induce its proteasome-mediated degradation (DC50 <10 nM), inhibit cancer

cell proliferation (EC50 <10 nM), have high in vivo exposure, and display a PD response in

vivo (Fig. 6). Optimized b-catenin degraders may thus become a first-in-class therapeutic to

address an unmet need in colon and other WNT-driven cancers.

 

MCL-1 (available for partnering)

MCL-1 is a member of the Bcl-2 family of proteins that binds to pro-death members of the

same family and inhibits apoptosis. It is overexpressed in many cancers, allowing cancer

cells to avoid apoptosis. Indeed, preventing programmed cell death is one of the hallmarks

of cancer. Thus, MCL-1 is a well validated cancer target. However, like other anti-apoptotic

members of the Bcl-2 family, MCL-1 is considered difficult to drug. Dr. Fesik has a long

history of working with Abbott/Abbvie on the Bcl-2 proteins. Prior work has included solving

the first structure of Bcl-xL (21) as well as Bcl-xL when bound to a peptide derived from the

pro-death protein BAK (22), discovery of the first potent inhibitor of Bcl-2, Bcl-xL, and Bcl-w

(23) that entered clinical trials (navitoclax), and discovery of a Bcl-2 selective inhibitor

(venetoclax, VENCLEXTA) that is now being used to treat patients with CLL and AML. At

Vanderbilt, the Fesik lab has targeted MCL-1 which causes resistance to Bcl-2 and Bcl-xL

inhibitors as well as other anticancer therapies such as gemcitabine, vincristine, and

paclitaxel.

 

To identify starting points for discovering MCL-1 inhibitors, we used a fragment-based

screen, which identified two hits that bound to different sites on the protein. We then used a

fragment merging strategy to obtain a much more potent MCL-1 inhibitor (24) (Fig. 7). From

extensive optimization, we discovered highly selective picomolar MCL-1 inhibitors with

enhanced cellular potency, drug-like PK properties, and in vivo efficacy by IV and PO dosing

(25-30) (Fig. 7). Under a licensing agreement with our then partner, Boehringer Ingelheim,

the MCL-1 inhibitors were extensively profiled. Potent antitumor efficacy in heme

malignancies and lung xenograft models was achieved without significant toxicity. Our leads

exhibit a different PK profile (lower Vss, shorter t1/2) compared to other MCL-1 inhibitors

which may be advantageous to manage the potential cardiovascular issues seen by MCL-1

inhibitors in clinical trials.

 

E3 LIGASE LIGAND DISCOVERY (available for partnering)

More than 600 E3 ligases are known, but only a handful (primarily cereblon and VHL) have

been used for targeted protein degradation. Novel ligands for different E3 ligases may be of

value for several reasons: 1) overcoming resistance caused by mutations in an E3 ligase

that block the ability of PROTACS to function, 2) expanding the applicability of PROTACS

to target proteins that cannot be effectively degraded with known E3 ligases, and 3) reducing

the toxicities associated with non-specific inhibition or degradation of the target protein. One

particularly exciting application for new E3 ligases is the prospect of inducing degradation

only in certain locations or tissues. PROTACs built from ligands for E3 ligases with

expression in cancer but not in normal tissues might be used to degrade target proteins with

known tissue-specific toxicities and thus dramatically improve the therapeutic window.

To demonstrate the utility of the approach, we identified E3 ligases with limited protein

expression in normal tissues (Fig. 8). One example is KLHL12. PROTACs that recruit this

E3 ligase would be ideal to degrade proteins with toxicities that would otherwise limit their

use. Using a fragment-based screen, we first identified hits that bound weakly to KLHL12,

optimized binding to ~50 nM using iterative structure-based design from >140 X-ray

structures of co-complexes, and prepared PROTACS that degrade BRD4 and Bcl-xL (Fig. 8).

Degraders that recruit ligases with a restricted expression profile could reduce the toxicity

of BRD4 inhibitors, Bcl-xL inhibitors, and inhibitors of other proteins that are highly

efficacious but exhibit dose-limiting toxicities. We have identified hits in fragment-based

screens of other E3 ligases (VHL, FEM1B, CBL-c, and TRAF4) using state-of-the-art NMR

experiments (Fig. 9) and have determined the X-ray structures of hits bound to the

proteins to enable structure-based affinity optimization. We believe that fragment-based

methods are ideal for discovering small molecule ligands for E3 ligases and would be open to

screening our fragment library against other E3 ligases.

 

Viral

 

SARS CoV2 (available for partnering)

The Covid-19 pandemic caused by SARS-CoV-2, the present and future variants of this

virus that could emerge, and the potential for other coronaviruses to cause future outbreaks

highlight the continuing need for new antiviral drugs targeting critical proteins in the

coronavirus life cycle. Following host infection, SARS-CoV-2 translates its genome into two

polyproteins, which require subsequent cleavage by two cysteine proteases (main protease

and papain-like protease (PLPro )) to generate functionally active proteins. Although inhibitors

of the main protease have been developed, no inhibitors of PLPro have reached the clinic.

The high homology of PLPro across the coronavirus family makes PLPro an attractive drug

target to overcome drug-resistant variants or new emerging coronaviruses in future.

 

Using an NMR-based screen of an in-house 13,824-molecule fragment library, we identified

hits that bind to two sites on PLPro and characterized the binding modes at each site using

X-ray crystallography (31) (Fig. 10). Guided by X-ray structures of co-complexes, we have

improved the binding affinity > 3-orders of magnitude from the initial hit using fragment

growing to exhibit cellular antiviral activity. Unlike other reported PLPro inhibitors that are

analogs of GRL-0617 (an inhibitor of SARS-CoV), our inhibitors are structurally distinct and

novel. Current work is focused on further optimizing the cellular potency, pharmaceutical

properties, and in vivo activity with the goal of discovering a PLPro inhibitor that is suitable

for clinical development.

 

a-viruses (available for partnering)

The Chikungunya virus (CHIKV), Venezuelan equine encephalitis virus (VEEV), and Eastern

equine encephalitis virus (EEEV) are alphaviruses that cause diseases ranging from

debilitating arthritis to lethal encephalitis in humans. CHIKV has caused several major

outbreaks over the past 20 years, is highly infectious via the aerosol route, and is considered

a potential bioterror threats. However, there are no FDA-approved antivirals for treating any

alphavirus infection.

 

We have selected the CHIKV nsP2 cysteine protease to target due to its essential role in

alphavirus replication and its ability to convert the viral nonstructural polyprotein into

functional components. We have identified hits in a fragment-based screen that bind to

CHIKV nsp2 and are now in the process of determining the X-ray structures of these hits

when bound to the protein.

 

Longevity (available for partnering)

Another field of interest in our lab is longevity. We are at a very early stage identifying

potential drug targets that are suitable for fragment-based screens using NMR. We welcome

the collaboration with those that may have the same interest in this exciting area.

 

 

References

1. Shuker, S.B., Hajduk, P.J., Meadows, R.P. and Fesik, S.W. Discovering high-affinity

ligands for proteins: SAR by NMR. Science 274, 1531-1534 (1996).

2. Sun, Q., et. al., Discovery of small molecules that bind to KRAS and inhibit SOSmediated

activation. Angew. Chem. Int. Ed., 51, 6140-6143 (2012).

3. Kessler, D., et. al., Drugging an undruggable pocket on KRAS. Proc. Natl. Acad. Sci.

U.S.A. 116, 15823-15829 (2019).

4. Sun, Q., et. al., A method for the second-site screening of KRAS in the presence of

a covalently attached first-site ligand. J. Biomol. NMR 60, 11-14 2014.

5. Broker, J. et. al., Fragment optimization of reversible binding to the switch II pocket

on KRAS leads to a potent, in vivo active KRASG12C inhibitor. J. Med. Chem. 65,

14614-14629 (2022).

6. Kim, D. et. al., Pan-KRAS inhibitor disables oncogenic signalling and tumour growth.

Nature 619, 160-166 (2023).

7. Popow, J. et. al. bioRxiv (https://doi.org/10.1101/2023.10.24.563163)

8. Thomas, L.R. et. al. Interaction with WDR5 promotes target gene recognition and

tumorigenesis. Molecular Cell 58, 440-452 (2015).

9. Simon, S.C. et. al., Discovery of WD repeat-containing protein 5 (WDR5)-MYC

inhibitors using fragment-based methods and structure-based design. J. Med.

Chem. 63, 4315-4333 (2020).

10. Wang, F. et. al., Discovery of potent 2-aryl-6,7-didydro-5H-pyrrolo[1,2-a]imidazoles

as WDR5-WIN-Site inhibitors using fragment-based methods and structure-based

design. J. Med. Chem. (2018).

11. Macdonald, J.D. et al., Discovery and optimization og salicyclic acid-derived

sulfonamide inhibitors of the WD repeat-containing protein 5-MYC protein-protein

interaction. J. Med. Chem. 62, 11232-11259 (2019).

12. Aho, E.R. et. al., Displacement of WDR5 from chromatin by a WIN site inhibitor with

picomolar affinity. Cell Reports 26, 2916-2928 (2019).

13. Thomas, L.R. et. al. Interaction of the oncoprotein transcription factor MYC with its

chromatin cofactor WDR5 is essential for tumor maintenance. PNAS 116, 25261-

25268 (2019).

14. Bryan, A.F. et. al., WDR5 is a conserved regulator of protein synthesis gene

expression. Nucleic Acids Res. 48, 2924-2941 (2020).

15. Aho, E.R., Weissmiller, A.M., Fesik, S.W., and Tansey, W.P. Targeting WDR5: A

WINning anti-cancer strategy? Epigenetic Insights 12, 1-5 (2019).

16. Tian, J., et. al., Discovery and structure-based optimization of potent and selective

WDR5 inhibitors containing a dihydroisoquinoline bicyclic core. J. Med. Chem. 63,

656-675 (2020).

17. Teuscher, K. B. et. al. Discovery of potent orally bioavailable WD repeat domain 5

(WDR5) inhibitors using a pharmacophore-based optimization. J. Med. Chem. 65,

6287-6312 (2022).

18. Teuscher, K. B. et. al., Structure-based discovery of potent WD repeat domain 5

inhibitors that demonstrate efficacy and safety in preclinical animal models. PNAS

120, 1-12 (2023).

19. Garralda, E. et. al., MYC targeting by OMO-103 in solid tumors: a phase I trial. Nat.

Med. 30, 762-771 (2024).

20. Anastas, J. N.; Moon, R. T. Wnt signalling pathways as therapeutic targets in cancer

Nat Rev Cancer 13, 11-26 .(2013).

21. Muchmore, S.W. et. al., X-ray and NMR structure of human Bcl-XL, an inhibitor of

programmed cell death. Nature 381, 335-341 (1996).

22. Sattler, M. et. al., Structure of Bcl-XL/Bak peptide complex: Recognition between

regulators of apoptosis. Science 275, 983-986 (1997).

23. Oltersdorf T., et. al., An inhibitor of Bcl-2 family proteins induces regression of solid

tumors Nature 435, 677-681 (2005).

24. Friberg, A., et. al., Discovery of potent myeloid cell leukemia-1 (Mcl-1) inhibitors

using fragment-based methods and structure-based design. J. Med. Chem. 56, 15-

30 (2013).

25. Burke, J. P. et. al., Discovery of tricyclic indoles that potently inhibit Mcl-1 using

fragment-based methods and structure-based design. J. Med. Chem. 58, 3794-3805

(2015).

26. Pelz, N.F., et. al., Discovery of 2-indol-acylsulfonamide Myeloid Cell Leukemia 1

(Mcl-1) inhibitors using fragment-based methods. J. Med. Chem. 59, 2054-2066

(2016).

27. Lee, T. et. al., Discovery and biological characterization of potent Myeloid Cell

Leukemia-1 (Mcl-1) inhibitors. FEBS Lett. 591, 240-251 (2017).

28. Shaw, S. et. al., Optimization of potent and selective tricyclic indole diazepinone

Myeloid Cell Leukemia-1 inhibitors using structure-based design. J. Med. Chem. 61,

2410-2421 (2018).

29. Lee, T. et. al., Discovery of potent Myeloid Cell Leukemia-1 (Mcl-1) inhibitors that

demonstrate in vivo activity in mouse xenograft models of human cancer. J. Med.

Chem. 62, 3971-3988 (2019).

30. Tarr, J.C., et. al., Discovery of potent Myeloid Cell Leukemia 1 (Mcl-1) inhibitor and

demonstration of in vivo activity in mouse xenograft models of hematological and

solid tumors. J. Med. Chem. (submitted for publication)

31. Taylor. A. J., et. al., Fragment-based screen of SARS-CoV-2 papain-like protease

(PLpro). ACS Med. Chem. Lett. (submitted for publication).