Mass Spectrometry-Based Solutions for Drugging the Undruggable
Whitepaper
Last Updated: July 25, 2024
(+ more)
Published: July 15, 2024
Credit: iStock
Proteomic technologies are revolutionizing the analytical toolbox available to support preclinical drug discovery.
Thanks to recent advances in workflows and instrumentation, mass spectrometry-based quantitative proteomics has been successfully applied for comprehensive proteome-scale profiling to support the discovery of drugs that target proteins that were assumed to be undruggable.
This whitepaper highlights new drug discovery platforms that can uncover and pharmacologically target previously undruggable proteins targets, making them accessible to therapeutic intervention by small-molecule drugs.
Download this whitepaper to explore:
- The discovery of covalent drugs using activity-based protein profiling
- The discovery of reversible drugs using high-throughput photoaffinity labeling mass spectrometry
- Case studies detailing protein inhibitor and small molecule binder discoveries
p. 1
Evotec’s Industrial-Scale
Mass Spectrometry-based
Proteomics Solutions for
Drugging the Undruggable
Evotec’s Industrial-Scale Mass Spectrometry-based Proteomics Solutions for Drugging the Undruggable
Author: Francois Autelitano, VP Translational Proteomics and Metabolomics
For further information:
Francois Autelitano
VP Translational Proteomics
and Metabolomics
Francois.Autelitano@evotec.com
Considering that current drugs mainly target proteins, proteomic technologies are revolutionizing the analytical toolbox available to support preclinical
drug discovery. Through recent advances in workflows and instrumentation,
mass spectrometry (MS)-based quantitative proteomics has been successfully applied for comprehensive proteome-scale profiling to support the discovery of drugs that target proteins that were assumed to be undruggable in
the past. Therefore, both pharmaceutical and biotech industries have sparked
a growing interest in molecular glues, targeted protein degraders (TPD),
targeted protein stabilizer (TPS), and targeted covalent ligands.
ScreenPep™ – Proteomics without Compromise
Evotec operates one of the largest high-end mass spectrometry facilities
worldwide for Next-Generation Proteomics research. With more than 25 years’
experience in mass spectrometry, chemical proteomics, and state-of-the-art
data analytics, Evotec offers a broad range of cutting-edge mass spectrometry-based proteomic technologies and services to address key issues in drug
and biomarker discovery. We continuously advance our capabilities in mass
spectrometry-based proteomics to ensure unrivalled comprehensiveness and
data quality when analyzing cells, animal models and patient samples.
Evotec’s ScreenPep™ platform drives the paradigm shift towards high-throughput proteomics profiling without making any compromise on coverage and
precision. Implementing fast automation and parallelization for processing of
samples across many instruments, we can scale up our throughput for largescale discovery proteomics to 300,000 samples per year or 800 samples per day.
ScreenPep™ is a single-shot Data Independent Acquisition (DIA) based
protein quantification workflow (1) offering ultra-deep proteome coverage
with reproducible and precise quantification of 10,000+ proteins per sample.
The ScreenPep™ workflow (Figure 1) is also extendable to post-translational
modifications including ubiquitination, phosphorylation, acetylation,
methylation, and S-nitrosylation.ScreenPep™ provides a scalable, tractable, fully automated
proteome-based pipeline in human cell lines for screening
small molecules that bind to or alter the abundance of
endogenous proteins, such as targeted protein degraders
and stabilizers.
Discovery of Covalent Drugs using HighThroughput Activity-Based Protein Profiling
Irreversible inhibitors are a powerful modality in drug
discovery. By forming a covalent bond with the protein
target, these inhibitors can exhibit great potency and
prolonged duration of action. In addition, irreversible
inhibitors offer other advantages, such as targeting
therapeutically relevant mutations or addressing difficult-to-drug targets. Despite these advantages, pharma
has historically avoided them because of the potential for
unpredictable off-target toxicities or immune responses. In the cases when pharma have proceeded with the
development of covalent compounds, compromises between reactivity, selectivity and potency have produced
safe and effective drugs.
The approval of multiple covalent drugs by the FDA in
the last years has prompted a resurgence of interest in
this class of compound. Key examples include covalent
tyrosine kinase inhibitors (TKIs) for cancer treatment,
including the Bruton’s tyrosine kinase (BTK) inhibitor
Ibrutinib (2 – 4), the epidermal growth factor receptor
(EGFR) inhibitor Osimertinib (5 – 7), and Sotorasib, a
RAS GTPase family inhibitor developed for the treatment
of solid tumours with KRAS G12C mutations (8 – 10).
In the discovery of novel covalent inhibitors, covalent
fragments (typical MW of less than 300 Da) provide a
vital toolbox. These fragments have several advantages
that can be summarized as follows: (i) excellent target
occupancy via covalent bond formation; (ii) selectivity towards distinct protein target residues (cysteine,
lysine, serine, …) using chemically diverse electrophilic
warheads (11 – 12); (iii) possibility to establish target
engagement and on-target mechanism of action using
mass spectrometry-based methods in parallel with target
residue mutagenesis; (iv) ability to pharmacologically
target pockets within disordered regions and cryptic
binding sites beyond the substrate pockets, thus allowing
the discovery of new allosteric drugs or ligands for targets
that lack well-defined binding pockets; (v) discovery of
potentially unknown binding sites. At Evotec, several
strategies have been developed to discover, develop, and
validate covalent fragments as a promising path to covalent ligand discovery.
Intact protein liquid chromatography mass spectrometry
(LC-MS) is a widely used target-based covalent fragment
screening strategy (13 – 15). Intact protein LC-MS can provide information on the accurate mass of a protein (and
proteoforms) alone or conjugated with a covalent fragment, and the relative abundance of the different species
across different conditions. Intact protein LC-MS provides
stoichiometry of covalent protein–fragment complex
formation by comparing the calculated mass difference
across vehicle control to fragment-treated samples. We
use high-throughput intact protein LC-MS to screen covalent fragment libraries against pure recombinant protein
constructs and identify the fragments hits and quantify
their labeling. Subsequent steps to the primary screen can
include biochemical assays, surface-plasmon resonance
(SPR) (16), and intact protein LC-MS to deconvolute the
reversible and irreversible components of the interactions
and the kinetic rate constants (kon and koff and thus the KD,
k
inact/KI) of each step. These parameters are particularly
useful to rank order compound hits by potency and help
medicinal chemistry teams better understand and optimize structure-activity relationship (SAR). To localize the
site of modification, Peptide Mass-Fingerprinting (PMF)
using LC-MS/MS is applied. Finally, this data can be
paired with crystal structures that fully characterize the
modification sites and guide the subsequent optimization
to therapeutic leads (15 – 17).
Evotec’s Industrial-Scale Mass Spectrometry-based Proteomics Solutions for Drugging the Undruggable p. 2
Cell Treatment
Human cells
Lyse
Red/Alk
Digest
Clean-up
Compounds
Sample Preparation DIA-MS Target
Identification and
Quantification
Figure 1: Overview of the ScreenPep™ platformFigure 2: Overview of the HT-ABPP platform
Treat
Lyse
Combine
Probe
Red/Alk
Digest
Clean-up
Enrich
DIA-MS Target
Identification and
Quantification
Human cells
Covalent library
% occupancy
Compounds
A more powerful approach is the application of bottom-up mass spectrometry-based proteomics to peptide
mapping on a larger scale. The basic workflow involves
enzymatic digestion of a target protein modified by a
covalent fragment, followed by the analysis of the resulting peptides via tandem mass spectrometry (LC-MS/
MS) and peptide fragment data analysis (18 – 19). This
approach can resolve each amino acid of the peptide
sequence allowing unbiased identification of the targeted
residue with site occupancy information.
Although target-based electrophilic library screens using
intact protein LC-MS is effective and relatively easy to
implement, it requires production and isolation of stable
recombinant target proteins. For many targets of interest
like multi-pass membrane proteins (e.g., G-proteincoupled receptors and multi-subunit receptors or ion
channels), this is either not feasible on a scale needed for
these experiments or impossible given intrinsic instability of the target. Moreover, in many cases, selectivity
against isolated protein constructs does not directly
translate into global chemoselectivity in a cellular
environment.
To address this, we have established a state-of-art
mass-spectrometry based chemoproteomic screening
platform with two main objectives: 1) discover previously unknown target sites within disordered regions,
cryptic or shallow binding pockets, and allosteric pockets on the surface of proteins 2) identify small molecule
fragments that interact in a highly selective manner
with those pockets directly in complex biological systems (cell/tissue lysates, live cells, or live organisms).
Evotec’s proteome-wide chemoproteomic screening
Evotec’s Industrial-Scale Mass Spectrometry-based Proteomics Solutions for Drugging the Undruggable p. 3
platform provides selectivity and target engagement
measurements that delivers a map of ligandable sites
across the entire proteome. This serves as a starting
point for understanding the druggability of potential
therapeutic targets and for the development of small
molecules against any class of protein in their native
cellular context.
Our platform leverages residue-based probes (RBPs)
that possess broadly reactive covalent warheads developed to modify side chains on the targeted amino acid
residue, including cysteine (20 – 21), lysine (22 – 23),
tyrosine (24 – 25), and aspartate/glutamate residues
(26). RBPs also bear a reporter group, such as a fluorophore (i.e. TAMRA) or an affinity tag (i.e. desthiobiotin)
for identification and enrichment purposes. Alternatively, a bioorthogonal chemical handle such as an
alkyne or azide can be installed on the probe, allowing
the attachment of reporter or affinity tags after the
protein target conjugation step for subsequent click
chemistry-enabled applications. Our High-Throughput
Activity-Based Protein Profiling (HT-ABPP) platform
combines a robotic liquid handling platform for enrichment and peptide clean-up, stable isotope labeling by
amino acids in cell culture (SILAC), and data-independent acquisition (DIA)-mass spectrometry (MS)-based
proteomic analysis (Figure 2). The platform allows
the consistent mapping of 60,000+ reactive cysteine
sites on 13,000+ unique proteins, and 12,000+ distinct
reactive lysine sites on 3,500+ unique proteins, across
many different human cell lines and primary cells with
a throughput of up to 100 samples per day. Mapping of
reactive tyrosine and Asp/Glu sites in various cell lines
is currently in progress.We have used this platform to screen diverse covalent
reactive libraries in live cells to identify new chemical
matter targeting proteins or protein families of interest. This strategy enables the assessment of cysteine
reactivity and ligandability directly in a cellular context.
Using dose-dependent competition assays in combination with quantitative MS, we can determine IC50 values
and apparent dissociation constants (Ki app) of the
electrophilic covalent fragments against all specifically
captured proteins in a single experiment (27 – 29). This
information enables rapid identification of covalent
ligands against reactive hotspots, along with providing
selectivity information on each ligand in various cellular
states. When paired with a parallel phenotypic screen
against a desired outcome, this methodology facilitates
the identification of functional covalent ligands and
their corresponding protein targets in a high-throughput manner.
Discovery of Reversible Drugs using
High-Throughput PhotoAffinity Labeling
Mass Spectrometry
The HT-ABPP approach can identify protein targets featuring suitably reactive nucleophilic (primarily cysteine
or lysine) residues. Since this significantly narrows the
target space, we developed an integrated photoaffinity
labeling mass spectrometry screening platform to test
Fully Functionalized Fragment (FFF) libraries in the
presence of purified protein (30) or live cells (31 – 32).
Each FFF probe contains a variable small-molecule
fragment conjugated to (i) a photoactivatable diazirine group, which is embedded within its structure to
permit UV light-induced cross-linking to interacting
proteins, and (ii) a click chemistry derivatizable handle
allowing the attachment of reporter or affinity tags for
Evotec’s Industrial-Scale Mass Spectrometry-based Proteomics Solutions for Drugging the Undruggable p. 4
subsequent visualization, enrichment, and identification of interacting proteins.
Using our High-Throughput PhotoAffinity Labeling
Mass Spectrometry (HT-PALMS) platform in combination with FFF libraries, we can globally map thousands
of reversible small-molecule fragment-protein interactions directly in human cells (Figure 3). In follow-up
studies, we can map the sites of protein binding for FFF
hits. Furthermore, FFF hits can immediately be used as
reporters in competition assays to screen focused libraries of structurally elaborated underivatized fragment
analogs (where the constant photoreactive/clickable
component is replaced with a propanamide group) to
establish SAR and identify more potent and selective
cell-active binders targeting protein(s) of interest.
Competition studies can then eloquently discern phenotypically relevant target(s) of FFF hits from proteins
that interact with the compounds but do not contribute
to the phenotype. From the elaborated competitor hits, a
second-generation library of FFF probes can be generated, expanded, and screened in live cells, as described
previously, to identify more potent FFF probes in a
site-specific manner.
Using our HT-PALMS platform to screen FFF libraries
in the presence of a purified protein in a biochemical
setting improves the throughput and enables targeted
screening against proteins of interest. FFF hits can be
detected by intact protein LC-MS, enabling identification of known and new chemotypes. FFF hits can be profiled to determine potency and the site of crosslinking,
and subsequently developed as reporters in a competitive displacement assay to identify novel starting
points in the development of therapeutics.
Treat
UV
Lyse
Click
Red/Alk
Enrich
Digest
Clean-up
Elaborated
fragment library
FFF probe
DIA-MS Target
Identification and
Quantification
Figure 3: Overview of the HT-PALMS platformEvotec’s Industrial-Scale Mass Spectrometry-based Proteomics Solutions for Drugging the Undruggable p. 5
Evotec’s Proprietary Covalent Libraries –
Covering diverse warheads and
Chemical Space
Evotec created an extensive collection of 5,300 acrylamide derivatives as cysteine modifiers (Figure 4). First
focus was on acrylamides because of (i) the growing interest in selective covalent modification of cysteine residues, (ii) the increasing number of acrylamide-derived
drugs reaching advanced clinical phases and the market
[Ibrutinib (2 - 4), Osimertinib (5 – 7), Sotorasib (8 – 10)],
(iii) our internal expertise built over years showing
developability of such molecules and (iv) their long-term
stability (over years), as demonstrated by their storage
in our sample management facilities. This collection
covers fragment and lead-like chemical spaces and was
designed using a thorough molecular design workflow
based on well-thought molecular descriptors, Evotec list
of structural alerts, and diversity selection. The outcome
of the workflow was supervised by skilled medicinal
chemists experienced in covalent drug discovery.
Figure 4: Evotec’s acrylamide-focused electrophilic fragment library.
Selection workflow (A) and main descriptors (B) of the Evotec acrylamide screening collection.
(A) (B) Finely-tuned acrylamide library
Long term storage, rapid compound progression, proven
development potential
Curation using an in-house database of frequent hitters and structural alerts
Relevant chemical property distribution
Visual inspection by seasoned Medicinal chemists
Representative examples
Distribution of molecular properties
~50k set
Final 5k set
Pooling of commercially available covalent
compounds from trusted vendors
Evotec Structural Alerts
Molecular descriptors
Confirmation of availability
Retrosynthetic Tractability Scoring
Diverse Subset Selection
Eye-check of remaining
structures
Evotec is now extending the collection towards other
protein residues that are susceptible to chemical
modification, such as lysine, serine/threonine, tyrosine,
aspartic/glutamic acids, methionine, arginine, and
histidine. We pay particular attention to stability when
creating new library sets, which has particularly high
importance when dealing with reactive molecules.
Hence our decision to test mid-term stability of islets of
relevant chemical modifiers of various protein residues
in regular storage conditions. Warheads that appear to
be suitable for mid-term storage will be extended to
create the “beyond cysteine” Evotec covalent collection.
Evotec also uses the libraries of covalent compounds and
FFFs commercially available from different suppliers
(e.g. Enamine, Life Chemicals, Molport) as well as the
libraries built up by his partners.Evotec’s Industrial-Scale Mass Spectrometry-based Proteomics Solutions for Drugging the Undruggable p. 6
Conclusion
We have established new drug discovery platforms that leverage groundbreaking
scientific approaches in MS-based proteomics to uncover and pharmacologically
target previously undruggable proteins targets, making them accessible to therapeutic
intervention by small-molecule drugs.
With the SreenPep™ platform, Evotec operates MS-based proteomics at industrial scale,
without compromise on sensitivity, precision, or proteome coverage, to screen extensive
small-molecule libraries for TPD of pathogenic proteins previously considered undruggable.
Significant advances in fragment-based screening, including the emergence of FFFs and
innovations in covalent fragment screening provide a new paradigm for ligand and target
discovery. Evotec’s HT-PALMS platform has the potential to fill major gaps in smallmolecule probe development by enabling the discovery of reversible ligands, and the sites
of ligand binding, for many proteins in parallel, directly in human cells.
Screening covalent fragments using our HT-ABPP platform enables exploration of
novel druggable pockets through irreversible fragment-amino acid residue interactions,
complementing their fully functionalized counterparts.
FFFs and covalent fragments are not only useful for identifying functional modulators,
but also play important roles in the emerging induced proximity modalities, including
molecular glues and rationally designed bifunctional agents. HT-PALMS and HT-ABPP
platforms facilitate the expansion of TPD and TPS approaches by enabling the discovery
of new E3 ubiquitin ligase or deubiquitinase recruiters.
With ScreenPep™, intact protein LC-MS, HT-PALMS, and HT-ABPP platforms, Evotec
provides the screening community with multiple unbiased and complementary routes
for discovering disease-focused targets and new chemical entities toward first-in-class
therapeutics.Case study 1: Discovery of selenocysteineselective covalent inhibitors of a
SelenoTarget protein
Rational
Selectively targeting tumors with drugs that induce
reactive oxygen species (ROS) production is a promising
and well-studied approach for cancer treatment, with the
promise to succeed in a clinical setting. Selenium (Se)
is one of the trace elements that is essential for human
survival. Its essential benefits to human health are mainly
due to its presence in proteins as selenocysteine (33).
The main role of Se is participating in the redox catalytic
process (34). Many selenoproteins have been found to
be associated with the occurrence and poor prognosis of
cancer (35 –36). In this study, we were interested in an
Se-related enzyme whose role in cancer is established that
we here call SelenoTarget (SeTarget). SeTarget inhibition
rapidly induces high ROS-levels in cancer cells, resulting
in cell death. Inhibition of SeTarget may thus contribute to
cancer therapy and improving chemotherapeutic agents.
Mammalian SeTarget contains a highly reactive, surface-exposed selenocysteine residue (Se-Cys or U) in its
active site. Selenocysteine is easily targeted by electrophilic
compounds that inhibit SeTarget activity. Herein, we
decribe a strategy to discover novel chemical matter that
inhibit the enzymatic activity of SeTarget through covalent
modification of the C-terminal selenocysteine (Figure 5).
Evotec’s Industrial-Scale Mass Spectrometry-based Proteomics Solutions for Drugging the Undruggable p. 7
Electrophile-focused library
We compiled a library of 1,047 electrophilic fragments
featuring mildly reactive cysteine-targeting warheads,
including chloroacetamide- (733) and acrylamide-based
(229) warheads, with average molecular weights of 241
and 233 Da, respectively, as well as heterocyclic electrophiles (85) with various types of warheads (e.g. haloalkyl,
vinyl sulfone, aldehyde ketone, and cyano groups) and an
average molecular weight of 413 Da. To this collection, we
added 95 electrophiles (average molecular weight of 413
Da and armed with α,β-unsaturated amides or ketones or
cyano groups) selected by target-based virtual screening
of over 100,000 cysteine-focused covalent compounds
against two X-ray structures of the SeTarget of interest,
targeting its catalytic selenocysteine residue.
Intact protein LC-MS screening
Human SeTarget protein construct was incubated with
each fragment in singleton at 230 μM for 2 h at room
temperature. Following incubation, intact protein liquid
chromatography–mass spectrometry (LC–MS) was applied
to identify the fragment hits and quantify the percentage
of SeTarget labeling.
In total, 96 compounds covalently bound to SeTarget
protein with a Drug-Antibody Ratio (DAR* is defined as
the average number of compounds adducts attached to
a single SeTarget protein) between 1 (mono-adduct) and
2 (bis-adduct) were selected for a counter-screen on the
mutated SeTarget construct (U -> S). Of these, 59 hits were
shown to form a single covalent adduct with SeTarget
selenocysteine over other cysteines on the protein with
high selectivity (ΔDAR** ≥ 0.95).
Enzymatic activity and thiol reactivity
Among the selected hits, 8 and 23 were confirmed to
inhibit enzymatic activity of the purified enzyme by
an endpoint enzymatic assay with IC50 values between
2–10 µM and 10–30 µM, respectively. Selected substances
were tested for glutathione (GSH) reactivity using glutathione reactivity assay (37–38). While there was variability
in the intrinsic reactivity of the fragments toward thiols,
it was sufficiently small to allow the identification of hits
sufficiently mild to ensure that the main driver of protein
labeling is recognition rather than reactivity.
Hit Expansion phase
Next, six clusters were selected based on enzymatic activity
(IC50 < 10 µM), lipophilicity (cLogD7.4 ≤ 3.5), and GSH reactivity (t1/2 > 1 h), and advanced for hit expansion via medicinal chemistry. In total, 273 analogs were synthesized
and tested for their enzymatic activity and thiol reactivity.
Several compounds exhibiting low single digit micromolar
Figure 5: High-throughput screening cascade for the discovery
of SeTarget covalent inhibitors targeting the catalytic selenocysteine residue. Summary of electrophile screening cascade and
hit validation showing number of primary hits from the highthroughput screen and how these were prioritized and filtered.
1,142 electrophiles from
a pre-plated cysteinefocused fragment library
95 ligands selected by target-based
virtual screening of >100K cysteinefocused covalent compounds
96 hits
1 < DAR < 2
Intact protein LC-MS primary
screen on SeTarget wild-type
Intact protein LC-MS counterscreen
on SeTarget (U->S) mutant
Enzymatic assay
Glutathione reactivity assay
Hit expansion phase
273 analogs synthesized
In-cell ABPP
59 hits
ΔDAR ≥ 0.95
6 clusters
IC50 < 30 µM; t1/2 > 1 h
31 hits
IC50 < 30 µM
16 hits
IC50 < 10 µM; cLogD (7.4) ≤ 3.5; t1/2 > 1 h
14 hits
engaged SeTarget
Se-Cys at ≥ 94%SeTarget enzymatic potency were discovered. Among these
hits, 16 compounds with IC50 values below 10 µM, t1/2 > 1 h,
and cLogD7.4 ≤ 3.5 with molecular weight (Lipinsky) below
400 Da were assessed for target engagement and selectivity
using in-cell Activity-Based Protein Profiling (ABPP) (19–20).
Activity-based protein profiling in human cells
Ligandable cysteine and selenocysteine sites were profiled
using a generalized desthiobiotin iodoacetamide probe
(DB-IAA) to differentiate conjugated cysteine- and selenocysteine-containing peptides not bound by electrophiles.
NCI-H23 cells were incubated with DMSO or 100 µM electrophilic compound hits for 2 h at 37°C in culture medium.
After treatment, cysteines and Se-Cys that did not get covalently labeled with the compounds were then alkylated with
the DB-IAA probe. Each compound was analyzed in three
biological replicates. Cells were lyzed in the presence of urea
and the free cysteine and Se-Cys sites reduced with DTT
then alkylated with iodoacetamide (IAA). After digestion
with trypsin/LysC cocktail, LC-MS/MS was used to quantify
the peptides containing cysteines and Se-Cys modified with
DB-IAA. The final output is displayed as a fold change (FC,
compound/DMSO) for each electrophile, where an FC ≤ -2
(with an adjusted p-value < 5%) represents > 50% attenuation
of cysteine/Se-Cys probe alkylation at that site. Starting with
100 μg of sample input per condition, in-cell ABPP quantified > 24,500 cysteine sites including 8 Se-Cys for > 6,600
unique proteins, with 15,900 - 21,800 ligandable sites per
Evotec’s Industrial-Scale Mass Spectrometry-based Proteomics Solutions for Drugging the Undruggable p. 8
Figure 6: Target engagement and selectivity profiling of SeTarget inhibitors in NCI-H23 cells using competition ABPP.
Competition ABPP of NCI-H23 cells was performed with compounds (20 and 100 µM) for 2 h using single-shot label-free proteomics
by data-independent acquisition (DIA)-MS. Data is presented as the mean of biological replicated experiments (n = 3). The final
output is displayed as a fold change (FC, compound/DMSO) for each compound, where an FC ≤ -2 (with an adjusted p-value < 5%)
represents > 50% attenuation of cysteine/selenocysteine DBIAA probe alkylation at that site.
(A) Diagram showing the percentage of occupancy of SeTarget selenocysteine (U) as well as the total number of cysteine/
selenocysteine sites significantly engaged for each compound (A–L).
(B) Volcano plot showing Log2[Fold Change] and significance (-Log10[p-value]) between cysteine/selenocysteine sites enriched
by DBIAA probe from cells treated with compound K versus vehicle. Compound K (100 µM) significantly engaged SeTarget
selenocysteine (U) as well as 10 other reactive cysteines and 1 selenocysteine from 11 different proteins.
(A) (B)
condition. Testing of 16 hits at high concentration (100 µM)
in NCI-H23 cells confirmed that all but two compounds
engage the C-terminal Se-Cys of SeTarget protein at ≥ 94%
among other cysteine and Se-Cys sites (12 to > 4,300) from
other proteins, presumably due to "overdosing" in this
experiment (Figure 6A). Two compounds (C and H) tested
at a lower dose (20 µM) showed as dose-dependent behavior
with a better selectivity towards SeTarget catalytic Se-Cys
residue. Finally, the most interesting hit compound (K),
when used at the highest concentration engaged SeTarget
selenocysteine > 98% with an excellent selectivity, with only
10 other cysteines and 1 selenocysteine from 11 different
proteins liganded between 77% and 95% (Figure 6B).
In conclusion, this study highlights the potential of electrophile-fragment screening as a practical and efficient tool
for covalent-ligand discovery. HT-ABPP platform enables
screening of electrophile libraries, and present comprehensive reactive cysteine maps for > 6,600 proteins and
ligandability information across electrophiles in cells at low
micromolar concentrations.
*DAR = ∑((0 adduct * v%) + (1 adducts * w%) + (2 adducts
* x%) + (3 adduct * y%) + (4 adduct * z%) / ∑(v% + w%+ x%
+ y% + z%) where v, w, x, y and z is the % of 0, 1, 2, 3 and 4
adducts
**ΔDAR = DAR (wt form) – DAR (mutant form)Evotec’s Industrial-Scale Mass Spectrometry-based Proteomics Solutions for Drugging the Undruggable p. 9
Case study 2: Discovery of small molecule
binders of the E3 ubiquitin ligase Trim 47
using photoaffinity-labeling combined with
mass spectrometry-based proteomics.
Rational
Tripartite Motif 47 (TRIM47) functions as a RING-finger
E3 ubiquitin ligase. Here, we report a holistic photoaffinity-labeling mass spectrometry screening approach to
identify novel binders for TRIM47 as warheads for future
Trim47 PROteolysis TArgeting Chimeras (PROTACs) and
molecular glues development. In the present study, we
used 3 Fully Functionalized Fragment (FFF) probes with
each member possessing a variable small-molecule fragment conjugated to a constant tag bearing an alkyne and
photoactivatable diazirine group (39) (Figure 7A). For
FFF1 and FFF2 probes, the variable fragment groups had
an average molecular weight of approximately 180 Da.
The variable fragment of FFF1 probe is a benzylpiperazine
group found in many biologically active natural products
and clinically approved drugs, like Imatinib (Gleevec)
and Meclizine (Bonine) to name just a few. The variable
fragment structure of FFF2 probe was not disclosed for
confidentiality reasons. Finally, a “fragmentless” photoreactive probe bearing a methyl group, was used as a
negative probe (NP).
Gel-based profiling of FFF probes in human cells
We initially assessed the FFF probes using gel-based
profiling by treating HEK293T cells with each fragment
probe (20 or 200 µM, 30 min), followed by exposure to
UV light (20 min, 4°C), cell lysis, coupling to a tetramethylrhodamine (TAMRA)-azide tag using copper-catalyzed azide alkyne cycloaddition (CuAAC) chemistry
(40), and separation and visualization of fragment-modified proteins by SDS-PAGE coupled with in-gel fluorescence scanning. Despite the structural simplicity and
small size of the variable fragment groups, FFF1 and
FFF2 probes produced marked and differential concentration-dependent protein labeling in HEK293T cells
(Figure 7B). Negligible protein labeling was observed
in the absence of UV light (data not shown), indicating that the fragment-protein interactions correspond
to reversible binding events that were converted to
covalent adducts upon photolysis. Protein labeling with
NP probe was almost undetectable and comparable to
that obtained in the absence of probe, indicating that
the variable group of FFF probes is critical for protein
binding.
MS-based profiling of FFF probes in human cells
We next set out to globally map fragment-binding proteins
in HEK293T cells by quantitative chemical proteomics
following the general protocol shown in Figure 3. We compared FFF probes at 200 µM (30 min incubation) to control
probe NP in pairwise experiments using single-shot
label-free proteomics by data-independent acquisition
(DIA)-MS, where proteins enriched by the test FFF probe
over NP with a fold change (FC) > 2 and adj p-value < 5%
were designated as test probe targets. In aggregate, 2,295
protein targets were identified for FFF1 or FFF2 probes,
with both probes individually displaying a broad range
of protein enrichments (820 and 2,227 proteins, respectively). When tested at lower concentrations (20 µM), FFF
probes enriched significantly less protein targets (data not
shown), confirming that the extent of proteome engagement depends on probe concentration. The probe-versuscontrol comparisons showed that 752 target proteins
(33%) were significantly enriched with both FFF probes,
whereas a subset of target proteins exhibited exclusive
interactions with either FFF1 (68) or FFF2 (1,475) probes.
TRIM47 was significantly enriched with FFF2 probe over
NP probe (FC = 2.43, adj p-value = 4.23E-10) but not with
FFF1 probe (Figure 7C).
Reversible small molecule interactome mapping using
PALMS in human cells
To confirm engagement of Trim47 by FFF2 probe in
cells, we performed competition photoaffinity displacement assay using two structurally elaborated fragment
analogues, CP1 and CP2, lacking the photoreactive
warhead. HEK293T cells were treated for 30 min with
10 µM FFF2 or NP as a control. For competitive profiling,
a 10 min-pretreatment with 160 µM competitor was
followed by FFF2 probe treatment. After photolysis,
FFF-modified proteins were enriched and identified
as previously described. Candidates are designated as
proteins enriched by FFF2 probe over NP with an FC >2
and adj p-value < 5% and competed off by the competitor
with a FC > 2 and adj p-value < 5% over FFF2 probe. A
total of 10 and 28 competed protein targets of FFF2 were
mapped for CP1 and CP2, respectively (Figure 7D). The
overlap between CP1 and CP2 fragment targets remained
modest, with only three common proteins including
Trim47. We found Trim47 to be a primary target of FFF2
and the two competitors, as it is significantly enriched
over NP (FC = 2.94 and adj p-value = 3.71E-7) and this
binding could be blocked upon co-incubation with CP1
(FC = 2.98 and adj p-value = 2.59E-6) or CP2 (FC = 3.16
and adj p-value = 7.70E-7).Evotec’s Industrial-Scale Mass Spectrometry-based Proteomics Solutions for Drugging the Undruggable p. 10
Figure 7: Small molecule interactome mapping by photoaffinity labeling mass spectrometry identifies reversible binders for
Trim 47 in human cells
(A) Structures of FFF probes. The “constant” (containing the diazirine photoreactive group and clickable alkyne group) and “variable”
(consisting of small-molecule fragments) regions of FFF probes are shown in red and blue, respectively.
(B) FFF probe-protein interactions in cells. HEK293T cells were treated with indicated FFF probes (20 or 200 µM) for 30 min, followed
by photocrosslinking, lysis, CuAAC conjugation to a tetramethylrhodamine (TAMRA)-azide tag, separation by SDS-PAGE, and
visualization by in-gel fluorescence scanning and Coomassie Blue staining.
(C) Volcano plots showing relative protein enrichment values of FFF probes (200 µM) versus NP in HEK293T cells. FFF probes at
200 µM (30 min incubation) were compared to NP in pairwise experiments using single-shot label-free proteomics by dataindependent acquisition (DIA)-MS. Proteins enriched by the test FFF probe over NP with a fold change (FC) > 2 and adj pvalue < 5%
were designated as test probe targets (red circles). Data is presented as the mean of biological replicated experiments (n = 3).
(D) Chemical proteomic target engagement of CP1 and CP2 in HEK293T cells. The x-axis shows protein enrichment by active probe
FFF2 over NP (10 μM), while the y-axis shows protein competition in cells treated with FFF2 (10 μM) and DMSO or the designated
competitor analog (160 μM). Proteins that were significantly enriched by FFF2 over NP are plotted as gray circles. Proteins that
were significantly competed off by competitor are plotted as pink circles. Candidates (highlighted in red circles) are designated as
proteins enriched by FFF2 probe over NP with an FC >2 and adj p-value <5% and competed off by the competitor with a FC > 2
and adj p-value < 5% over FFF2 probe. Data is presented as the mean of biological replicated experiments (n = 3).
(A)
(D)
(B) (B)
(C)
In this work, we have shown that HT-PALMS platform
has the potential to map small-molecule fragmentprotein interactions directly in human cells. The case
study investigated herein on Trim47, for which selective
ligands were still lacking, underscores the versatility
and scope of PALMS approach for accelerating the
discovery of reversible small molecules for difficult
protein targets.References
1. Gillet LC, Navarro P, Tate S, Röst H, et al. Targeted Data Extraction of the MS/MS Spectra Generated by DataIndependent Acquisition: A New Concept for Consistent and Accurate Proteome Analysis. Mol Cell Proteomics.
2012 Jun;11(6):O111.016717.
2. Byrd JC, Furman RR, Coutre SE, Flinn IW, et al. Targeting BTK with Ibrutinib in Relapsed Chronic Lymphocytic
Leukemia. N Engl J Med. 2013 Jul 4;369(1):32-42.
3. Byrd JC, Furman RR, Coutre SE, Flinn IW, et al. Ibrutinib Treatment for First-Line and Relapsed/Refractory
Chronic Lymphocytic Leukemia: Final Analysis of the Pivotal Phase Ib/II PCYC-1102 Study. Clin Cancer Res.
2020 Aug 1;26(15):3918-3927.
4. Ahn IE, Brown JR. Targeting Bruton's Tyrosine Kinase in CLL. Front Immunol. 2021 Jun 23;12:687458.
5. Cross DAE, Ashton SE, Ghiorghiu S, Eberlein C et al. AZD9291, an Irreversible EGFR TKI, Overcomes T790MMediated Resistance to EGFR Inhibitors in Lung Cancer. Cancer Discov. 2014;4(9):1046–1061.
6. Shah R, Lester JF. Tyrosine kinase Inhibitors for the Treatment of EGFR Mutation-Positive Non-Small-Cell
Lung Cancer: A Clash of the Generations. Clin Lung Cancer. 2020;21:e216–e228.
7. Lamb YN, Scott LJ. Osimertinib: A Review in T790M-Positive Advanced Non-Small Cell Lung Cancer. Target Oncol.
2017;12(4):555–562.
8. Canon J, Rex K, Saiki AY, Mohr C, et al. The Clinical KRAS(G12C) Inhibitor AMG 510 Drives Anti-Tumour Immunity.
Nature. 2019 Nov;575(7781):217-223.
9. Lanman BA, Allen JR, Allen JG, Amegadzie AK, et al. Discovery of a Covalent Inhibitor of KRASG12C (AMG 510)
for the Treatment of Solid Tumors. J Med Chem. 2020 Jan 9;63(1):52-65.
10. Skoulidis F, Li BT, Dy GK, Price TJ, et al. Sotorasib for Lung Cancers with KRAS p.G12C Mutation. N Engl J Med.
2021 Jun 24;384(25):2371-2381.
11. Benns HJ, Wincott CJ, Tate EW, Child MA. Activity- and Reactivity-Based Proteomics: Recent Technological
Advances and Applications in Drug Discovery. Curr Opin Chem Biol. 2021 Feb;60:20-29.
12. Spradlin JN, Zhang E, Nomura DK. Reimagining Druggability Using Chemoproteomic Platforms. Acc Chem Res.
2021 Apr 6;54(7):1801-1813.
13. Resnick E, Bradley A, Gan J, Douangamath A, et al. Rapid Covalent-Probe Discovery by Electrophile-Fragment
Screening. J Am Chem Soc. 2019 Jun 5;141(22):8951-8968.
14. Dubiella C, Pinch BJ, Koikawa K, Zaidman D, et al. Sulfopin Is a Covalent Inhibitor of Pin1 that Blocks Myc-Driven
Tumors in Vivo. Nat Chem Biol. 2021 Sep;17(9):954-963.
15. Douangamath A, Fearon D, Gehrtz P, Krojer T, et al. Crystallographic and Electrophilic Fragment Screening of
the SARS-CoV-2 Main Protease. Nat Commun. 2020 Oct 7;11(1):5047.
16. Gunnarsson A, Christopher J, Stubbs CJ, Rawlins PB, Taylor-Newman E, et al. Regenerable Biosensors for
Small-Molecule Kinetic Characterization Using SPR. SLAS Discov. 2021 Jun;26(5):730-739
17. Davies DR. Screening Ligands by X-ray Crystallography. Methods Mol Biol. 2014;1140:315-23.
Evotec’s Industrial-Scale Mass Spectrometry-based Proteomics Solutions for Drugging the Undruggable p. 1118. Boike L, Cioffi AG, Majewski FC, Co J, et al. Discovery of a Functional Covalent Ligand Targeting an Intrinsically
Disordered Cysteine Within MYC. Cell Chem Biol. 2021 Jan 21;28(1):4-13.e17.
19. Al-Khawaldeh I, Al Yasiri MJ, Aldred GG, Basmadjian C, et al. An Alkynylpyrimidine-Based Covalent Inhibitor That
Targets a Unique Cysteine in NF-κB-Inducing Kinase. J Med Chem. 2021 Jul 22;64(14):10001-10018.
20. Vinogradova EV, Zhang X, Remillard D, Lazar DC, et al. An Activity-Guided Map of Electrophile-Cysteine
Interactions in Primary Human T Cells. Cell. 2020 Aug 20;182(4):1009-1026.e29.
21. Kuljanin M, Mitchell DC, Schweppe DK, Gikandi AS, et al. Reimagining High-Throughput Profiling of Reactive
Cysteines for Cell-Based Screening of Large Electrophile Libraries. Nat Biotechnol. 2021 May;39(5):630-641.
22. Hacker SM, Backus KM, Lazear MR, Forli S, et al. Global Profiling of Lysine Reactivity and Ligandability in the
Human Proteome. Nat Chem. 2017 Dec;9(12):1181-1190.
23. Abbasov ME, Kavanagh ME, Ichu TA, Lazear MR, et al. A Proteome-Wide Atlas of Lysine-Reactive Chemistry.
Nat Chem. 2021 Nov;13(11):1081-1092.
24. Hahm HS, Toroitich EK, Borne AL, Brulet JW, et al. Global Targeting of Functional Tyrosines Using Sulfur-Triazole
Exchange Chemistry. Nat Chem Biol. 2020 Feb;16(2):150-159
25. Borne AL, Brulet JW, Yuan K, Hsu KL. Development and Biological Applications of Sulfur-Triazole Exchange
(SuTEx) Chemistry. RSC Chem Biol. 2021 Apr 1;2(2):322-337.
26. Ma N, Hu J, Zhang ZM, Liu W, et al. 2H-Azirine-Based Reagents for Chemoselective Bioconjugation at Carboxyl
Residues Inside Live Cells. J Am Chem Soc. 2020 Apr 1;142(13):6051-6059.
27. Miyahisa I, Sameshima T, Hixon MS. Rapid Determination of the Specificity Constant of Irreversible Inhibitors
(kinact/KI) by Means of an Endpoint Competition Assay. Angew Chem Int Ed Engl. 2015 Nov 16;54(47):14099-102.
28. Strelow JM. A Perspective on the Kinetics of Covalent and Irreversible Inhibition. SLAS Discov. 2017 Jan;22(1):3-20.
29. Thorarensen A, Balbo P, Banker ME, Czerwinski RM, et al. The advantages of Describing Covalent Inhibitor In Vitro
Potencies by IC 50 at a Fixed Time Point. IC 50 Determination of Covalent Inhibitors Provides Meaningful Data to
Medicinal Chemistry for SAR Optimization. Bioorg Med Chem. 2021 Jan 1;29:115865.
30. Grant EK, Fallon DJ, Hann MM, Fantom KGM, et al. A Photoaffinity-Based Fragment-Screening Platform for
Efficient Identification of Protein Ligands. Angew Chem Int Ed Engl. 2020 Nov 16;59(47):21096-21105.
31. Parker CG, Galmozzi A, Wang Y, Correia BE, et al. Ligand and Target Discovery by Fragment-Based Screening
in Human Cells. Cell. 2017 Jan 26;168(3):527-541.e29.
32. Wang Y, Dix MM, Bianco G, Remsberg JR, et al. Expedited Mapping of the Ligandable Proteome Using Fully
Functionalized Enantiomeric Probe Pairs. Nat Chem. 2019 Dec;11(12):1113-1123.
33. Labunskyy VM, Hatfield DL, Gladyshev VN. Selenoproteins: Molecular Pathways and Physiological Roles.
Physiol Rev. 2014;94(3):739–77.
34. Rahmanto AS, Davies MJ. Selenium-Containing Amino Acids as Direct and Indirect Antioxidants.
IUBMB Life. 2012;64(11):863–71.
35. Hatfield DL, Tsuji PA, Carlson BA, Gladyshev VN. Selenium and Selenocysteine: Roles in Cancer, Health,
and Development. Trends Biochem Sci. 2014;39(3):112–20.7
Evotec’s Industrial-Scale Mass Spectrometry-based Proteomics Solutions for Drugging the Undruggable p. 1236. Tan HW, Mo H-Y, Lau AT, Xu Y-M. Selenium Species: Current Status and Potentials in Cancer Prevention and
Therapy. Int J Mol Sci. 2019;20(1):75.
37. Flanagan ME, Abramite JA, Anderson DP, Aulabaugh A, et al. Chemical and Computational Methods for the
Characterization of Covalent Reactive Groups for the Prospective Design of Irreversible Inhibitors. J Med Chem.
2014 Dec 11;57(23):10072-9.
38. Ábrányi-Balogh P, Petri L, Imre T, Szijj P, et al. A Road Map for Prioritizing Warheads for Cysteine Targeting Covalent
Inhibitors. Eur J Med Chem. 2018 Dec 5;160:94-107.
39. Li Z, Hao P, Li L, Tan CY, Cheng X, et al. Design and Synthesis of Minimalist Terminal Alkyne-Containing Diazirine
Photo-Crosslinkers and Their Incorporation Into Kinase Inhibitors for Cell- and Tissue-Based Proteome Profiling.
Angew Chem Int Ed Engl. 2013 Aug 12;52(33):8551-6.
40. Rostovtsev VV, Green LG, Fokin VV, Sharpless KB. A Stepwise Huisgen Cycloaddition Process: Copper(I)-Catalyzed
Regioselective "Ligation" of Azides and Terminal Alkynes. Angew Chem Int Ed Engl. 2002 Jul 15;41(14):2596-9.
Evotec’s Industrial-Scale Mass Spectrometry-based Proteomics Solutions for Drugging the Undruggable p. 13
2024/06 | V1
Brought to you by
Download the Whitepaper for FREE Now!
Information you provide will be shared with the sponsors for this content. Technology Networks or its sponsors may contact you to offer you content or products based on your interest in this topic. You may opt-out at any time.