Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

REvIEwS

Monitoring protein communities and


their responses to therapeutics
Hanna G. Budayeva and Donald S. Kirkpatrick ✉

Abstract | Most therapeutics are designed to alter the activities of proteins. From metabolic
enzymes to cell surface receptors, connecting the function of a protein to a cellular phenotype,
to the activity of a drug and to a clinical outcome represents key mechanistic milestones during
drug development. Yet, even for therapeutics with exquisite specificity, the sequence of events
following target engagement can be complex. Interconnected communities of structural,
metabolic and signalling proteins modulate diverse downstream effects that manifest as
interindividual differences in efficacy, adverse effects and resistance to therapy. Recent advances
in mass spectrometry proteomics have made it possible to decipher these complex relationships
and to understand how factors such as genotype, cell type, local environment and external
perturbations influence them. In this Review, we explore how proteomic technologies are
expanding our understanding of protein communities and their responses to large- and
small-molecule therapeutics.

A single human cell contains an estimated 10 billion be the case for a community of proteins expressed down-
individual proteins1. The molecules that comprise the stream of a transcription factor or the enzymes involved
cellular proteome derive from the mRNA expression of in a broad-acting cellular process (for example, protein
approximately 12 thousand distinct genes2–6 and range in degradation). Integrating these concepts, we define
abundance from one to upward of 10 million copies per a protein community as a set of proteins within a cell
cell7. Protein molecules may exist within a subcellular that are interconnected through one or more physical,
compartment or may transit between compartments as spatial, co-regulatory or functional relationships.
part of their dedicated functions (Fig. 1). Like a citizen The composition and dynamics of protein communi-
residing in a crowded metropolis, a protein molecule will ties are controlled by integrated regulation at the DNA,
encounter innumerable others during its lifetime. RNA and protein levels (Fig. 2). The genome of a cell,
When considering the state of a cell and its response its epigenetic state and the corresponding gene expres-
to therapeutic agents, the concept of protein com- sion profile define the repertoire of proteins that may
munities provides a valuable framework for forming be present. Downstream of gene transcription, mRNA
hypotheses and interrogating data. The largest protein splicing and translation further shape the emergence
community within an organism is its proteome — the of proteins, whereas lysosomal proteolysis, proteaso-
superset of all proteins that may be expressed in a given mal degradation and secretion maintain proteostasis.
cell. If we zoom in to the cellular level, a proteome can be Amongst the available mechanisms, protein–protein
envisaged as a multidimensional array of protein com- interactions (PPIs) frequently control the activity, sta-
munities related through physical interactions, subcel- bility and localization of proteins within a community.
lular colocalization (spatial communities), coordinated Similarly, post-translational modifications (PTMs) con-
regulation (co-regulatory communities) or biochemical trol protein activity, stability and localization, in addi-
activity (functional communities) (Fig. 2). Just as with tion to modulating PPIs. From common marks, such
human communities, the members of a protein com- as phosphorylation, acetylation and ubiquitylation, to
munity may physically interact, or they could equally rare or cell-type-specific events like hypusinylation8 or
well reside passively in the same subcellular location. crotonylation9, the complexity imparted to the proteome
Department of Microchemistry, Certain functional protein communities are defined as by PTMs is immense. Conditionally co-regulated PPIs
Proteomics and Lipidomics,
Genentech, South San
the recipients of input from a common source, as in the and PTMs represent key dimensions with which to
Francisco, CA, USA. case of phosphorylation substrates downstream of an define protein communities.
✉e-mail: donaldk@gene.com activated kinase. In other cases, proteins from a func- Questions involving where proteins reside within
https://doi.org/10.1038/ tional community may be connected not by proximity cells and which other proteins they physically inter-
s41573-020-0063-y or signalling, but rather by a common origin. This would act with, have long been of interest in proteomics and

Nature Reviews | Drug Discovery


Reviews

Stromal cell

EGF
a Analysing activation
versus inhibition of
EGFR signal transduction

Cancer cell P P Ras


P P P P
Endosome
RAF
d Studying b Studying
changes in biochemical
protein location c Analysing changes inhibition
in protein levels MEK P
Ub Ub Lysosome
Ub
Ub
P
ERK

Golgi

ER

ETV Gene expression

Fig. 1 | Proteins function within multiple protein communities. Extracellular activation of a plasma membrane receptor
(such as epidermal growth factor receptor (EGFR)) initiates signalling events that propagate across multiple protein
communities within a cell. A phosphorylation-mediated signalling pathway delivers the message to the nucleus, where
the transcriptional machinery controls downstream gene expression. The receptor can then redistribute across multiple
cellular compartments: endoplasmic reticulum (ER) and Golgi trafficking regulate folding and delivery of the receptor
to the plasma membrane, whereas ubiquitylation can initiate endosomal recycling or lysosomal degradation. In a tissue,
cell-to-cell communication and changes in the extracellular environment result in the initiation of cell-specific signalling
cascades. Comparing signal transduction downstream of a receptor that results from either activation or inhibition —
by, for example, antibody-based therapeutics (a) — can elucidate the members of physical, spatial, co-regulatory and
functional protein communities. Studying perturbations in signalling following biochemical inhibition (b), as well as
profiling drug–protein interactions, can help elucidate the drug mechanism of action. Changes in global protein levels
can indicate on- and off-target effects of molecules that trigger inducible protein degradation (c). Proximity-labelling
approaches and proteomic analysis of organelle fractions can determine changes in protein localization (d), such as
receptor endocytosis and recycling to the plasma membrane.

cell biology. Increasingly, now, this is also the case in As drug development progresses against the next
drug development. Cell surface proteins can only sense generation of challenging targets, understanding the
extracellular signals directly if they are on the plasma interconnected protein communities that they partici-
membrane. Transcription factors only drive RNA pate in is critical. This Review covers the growing role
expression if they are present in the nucleus. Likewise, of mass spectrometry proteomics in defining the phys-
multicomponent enzyme complexes must come together ical, spatial, co-regulatory and functional relationships
and engage their substrates if they are to control cellu- of proteins, the ways in which therapeutics affect protein
lar processes. An emerging body of evidence points to communities, and the value of these experiments in drug
‘non-canonical’ functions of certain organelle-specific development.
proteins when they become localized to different com-
partments10,11. As such, tumour-promoting functions of Advancements in proteomic technologies
metabolic enzymes that are independent of their cata- Steady improvements over the past two decades in
lytic activities may be targeted through modulation of analytical mass spectrometry and protein biochemis-
their specific subcellular localizations (reviewed in11). try have propelled the proteomics field by facilitating
Defining the community of proteins that reside at the the characterization of cellular and tissue proteomes
synapse of a neuron12, the identities of receptors on a in great detail15 (Fig. 3; Table 1). Advances in the speed
brain endothelial cell13 or the subcellular distribution of and sensitivity of mass spectrometry instrumentation,
oncogenic signalling modules in a cancer cell14 can be along with requisite improvements in downstream data
early steps in establishing novel therapeutic targets. analy­sis pipelines, have been at the heart of this progress.

www.nature.com/nrd
Reviews

Spectral counting
Together, these developments make it feasible to survey localization for small numbers of proteins in situ38.
Quantitative analysis based on most of the proteome in a matter of hours3,6,16. Even Protein quantification across conditions was at the core
comparing the total number of the more neglected regions of the proteome can now of the initial mechanistic studies for communities of
spectra identified per protein be explored using alternative proteolytic enzymes17 or proteins, including receptor tyrosine kinases (RTKs) and
in a mass spectrometry
enrichment protocols. For PTMs such as phosphoryl- G protein-coupled receptors (GPCRs), laying the foun-
analysis.
ation18,19, acetylation20,21, ubiquitylation22,23 and other dation for proteome-profiling studies with applications
Multiplexed isobaric processes, peptide enrichment strategies have yielded to drug discovery.
tagging extensive catalogues of marks that can occur on each Although much has been learned from sequencing
Mass spectrometry (MS)-based
protein24. These approaches extend to features such as the technologies, the characterization of DNA and RNA, by
quantification approach, in
which labelled peptides from
reactive cysteines, which convey information about definition, cannot provide information about the phys-
multiple samples are mixed the activity of proteins or their availability for engaging ical, spatial or functional relationships of most protein
together and analysed by mass with a ligand25,26. Whereas many PTMs occur at low fre- communities. Systematic studies that compare mRNA
spectrometer. Upon peptide quency and require specialized enrichment in order to and protein expression profiles have shown that gene
fragmentation, reporter ions of
be assessed, recent advances in instrument performance expression holds only partial regulatory control over
different masses are released
from the isobaric tags, and have opened the door to directly profiling certain modi­ protein levels4,39,40. Even for co-regulated gene sets, the
their relative intensities are fications6,27. As such, a recently reported shotgun pro- inducible degradation of a single component can desta-
measured within individual teomic workflow that did not include PTM enrichment bilize the remaining complex subunits, rendering gene
MS2/MS3 scan events. resulted in the identification of 7,000 N-acetylation expression futile. As drug development moves headlong
Chemoproteomics
and 10,000 phosphorylation sites in HeLa cells6. In into the inducible degradation of disease-causing pro-
Proteomics approaches that parallel, high-throughput studies evaluating thou- teins41, understanding the relationships between protein
use chemical probes to sands of proteins as baits in immunoaffinity pull-down communities becomes particularly important.
elucidate small-molecule– experiments have driven systematic cataloguing of
protein interactions in the
the physi­cal relationships between constituents of the Targeting protein communities
proteome.
human proteome28,29. One such study revealed more Mechanistic studies that involve therapeutic agents and
Proximity labelling than 1,300 protein communities that display physical their targets have revealed many important tenets con-
Proteomics approaches that (and presumably spatial and functional) relationships cerning the dynamic organization of cellular protein
use genetically engineered
upon overexpression in cultured human cells28. communities as controlled by PTMs and PPIs. On the
enzymes for the labelling and
enrichment of proteins within
Alongside improvements in sample handling and drug development side, model systems used to study
subcellular regions and protein analytical mass spectrometry have come improved the biochemistry and cell biology of these communities
communities. methods to quantify proteins across many samples have been valuable in differentiating between early ther-
(Box 1). Notable amongst these are protocols for per- apeutic candidates. Building upon these foun­dations,
forming quantification based on spectral counting, inte- mass spectrometry proteomics is helping define the
grated peak areas or multiplexed isobaric tagging30. Global composition and dynamics of protein communities
proteome profiling, PTM profiling, activity-based relevant to drug development (Fig. 1; Table 1).
protein profiling (ABPP) and chemoproteomics studies RTKs and the proteins that they engage with have
have benefited from multiplexed quantification and have been central to building our understanding of how pro-
been used for characterizing the mechanisms of drug tein communities respond to therapeutic agents. RTKs
action21,31–33. Quantitative methods have also helped elu- localize to the plasma membrane and generally func-
cidate the organizational structure of cellular compart- tion as sensors of the extracellular environment (Fig. 1).
ments and the signalling modules in them34–37. Spatial Classically, engagement of an RTK with its ligand ini-
proteomic approaches that involve density centrifu- tiates autophosphorylation of the intracellular tyrosine
gation and proximity labelling provide data for a broad kinase domain, signal propagation through a commu-
range of proteins in a single analysis, and thus com- nity of effector molecules and, ultimately, modulation
plement imaging-based methods that examine protein of the expression of a target gene42,43. Signal duration

Input Proteome Proteomics Protein communities


Genotype Physical

Cell type Functional

Environment Spatial

Pertubation Co-regulatory

Fig. 2 | Protein communities from the prism of mass spectrometry-based proteomics. Genotype, cell type,
environment and extracellular perturbations define the state of the proteome and its communities at a given point in time.
Mass spectrometry-based proteomics can decipher protein relations within physical, functional, spatial and co-regulatory
protein communities.

Nature Reviews | Drug Discovery


Reviews

Liquid
Peptide chromatography Target protein
Cancer cell Protein mixtures Immunoaffinity resin mixtures mass spectrometer community

Affinity Lysis • Physical


purification interaction

Label

Affinity resin B
Proximity • Physical
Lysis B interaction
labelling
• Functional
B interaction
B

Fractionation Fractions
Organelle • Spatial
Digestion
proteome interaction
profiling • Co-regulatory
interaction

PTM affinity resin • Physical


PTM P P interaction
Lysis P Digestion P • Spatial
profiling
interaction
P P P • Co-regulatory
P
P interaction
P
Drug
Inhibitor resin

Chemoaffinity
enrichment Lysis • Functional
interaction

Soluble
fractions
Thermal
proteome Lysis Digestion
profiling • Functional
interaction
Temperature
curve

Fig. 3 | Proteomic approaches for basic biology and drug discovery. Immunoaffinity purification identifies protein
interactions upon antibody-mediated isolation of a target protein with its complexes. Proximity labelling in cells captures
transient and functional interactions, as well as subcellular colocalization, by labelling with a reactive substrate followed
by affinity purification (such as streptavidin-mediated isolation of biotin-labelled products). Organelle proteome profiling
identifies the spatial protein distribution and co-regulation of proteins enriched in organellar fractions. Post-translational
modification (PTM) profiling can indicate protein association, localization and co-regulation through the enrichment and
quantification of modified peptides (for example, phosphorylated peptides). Chemoaffinity enrichment uses chemical
inhibitors (such as kinase inhibitors) as an affinity matrix to profile biochemical activities in the presence or absence of
a drug (such as another kinase inhibitor of interest). The expression levels, activity and affinity of an isolated enzyme for
the drug can be inferred from such study. Thermal proteome profiling leverages the concept of protein thermal stability
to interrogate small-molecule binding to direct targets and the affected protein communities, united by a common
biochemical response to a drug.

is regulated by internalization of the activated recep- Two heavily studied representatives of the RTK com-
tors from the plasma membrane and, in some cases, by munity are the epidermal growth factor receptors EGFR
their degradation44. Activators and inhibitors of RTKs (encoded by EGFR) and HER2 (encoded by ERBB2)43.
reshape numerous proteins through controlling syn- Both proteins are mutated and overexpressed in a variety
thesis, activity, localization and stability. The dynamics of tumours and have been targeted by small-molecule
of both the initial and downstream processes depend inhibitors and therapeutic antibodies, such as lapatinib
greatly on the abundance of the RTKs, the composition and trastuzumab, respectively43,45. Proteomic technology
of the protein communities they engage with and the was applied early on to elucidation of the relationships
PTM states of community members at various stages. between members of the ERBB community44,46,47, as
By extension, the dynamics of signalling within the well as their responses to tyrosine kinase inhibition32.
interconnected RTK community can directly affect Mass spectrometry proteomics combined with path-
the efficacy of therapeutic agents (Fig. 1). way activation or inhibition has been used to elucidate

www.nature.com/nrd
Reviews

Phosphoproteomics
the protein communities downstream of many RTKs, receptor (ER)-positive breast cancer, overexpression of
Proteomics approaches used including SRC family kinases (SFKs)48, FLT3 (ref.49), the kinase AXL has been tied to acquired resistance to
for the characterization of MET50, BCR-ABL51 and vascular endothelial growth HER2-targeting therapeutics55, and in gastric cancer,
proteins post-translationally factor (VEGF) 52. Because compensatory signalling profiling of trastuzumab-resistant cancer cells revealed
modified with a phosphate
between these communities is frequently implicated in activation of the mTOR pathway and sensitivity to
group by kinase enzymes.
drug resistance, these studies provide a framework for mTOR kinase inhibition56. In the case of lung cancer
evaluating and developing novel combinations53. cells treated with the tyrosine kinase inhibitor dasat-
Another area in which proteomics-based methods inib, both chemoproteomics and phosphoproteomics
have proved valuable is in determining the mechanisms together revealed broad-spanning inhibitory responses,
of acquired resistance that occur through rewiring of manifested in decreased phosphorylation events
otherwise silent signalling nodes (Table 1). Rexer et al. amongst members of the kinase community and their
used phosphotyrosine profiling of lapatinib-resistant substrates57. Several putative targets of dasatinib were
breast cancer cell lines48 to reveal activation of SFKs, and identified, such as a community of kinases including
Boyer et al. demonstrated context-dependent changes non-RTKs (such as ABL and SFK family members),
in members of the SRC and MAPK pathways, as well as in RTKs (such as ephrin receptors and EGFR) and serine–
FAM83A in trastuzumab-resistant, HER2-amplified threonine kinases (such as G-associated kinase and the
breast cancer cell lines54. In HER2-positive, oestrogen QIK subfamily)57.

Table 1 | Applications and limitations of proteomic approaches for the characterization of protein communities
experimental approach community Applications in drug discovery Limitations
Affinity purification Physical and Identification of drug-dependent PPIs Tag-mediated purification requires bait tagging
spatial and functional validation
Identification of drug–protein interactions Endogenous protein purification requires
validated antibody reagents
Characterization of protein complex Artificial interactions can be introduced during
stoichiometry cell lysis
Proximity labelling Physical, spatial Characterization of unbounded cellular Technique requires bait tagging and functional
and functional compartments validation
Capture of transient PPIs Method frequently involves exogenous bait
expression. Alternatively, a proximity-labelling
enzyme can be inserted within the endogenous
gene locus via cellular engineering
Organelle proteome profiling Spatial and Characterization of protein communities Sample preparation is laborious
co-regulatory of all major organelles in one dataset
Identification of drug-induced translocation Technique requires advanced bioinformatics
events analysis in order to translate mass spectrometry
data into protein distribution between organelle
Identification of secreted signalling factors fractions
Global proteome profiling Co-regulatory Protein co-regulation in disease models or Method is limited by the wide dynamic range
upon drug treatment of protein abundances, which complicates
quantification of the lower abundance proteins
Chemoaffinity enrichment Functional Identification of potential drug targets and Technique may require optimization for drug
off-targets conjugation to the beads
Drug binding does not necessarily reflect activity
in cells
Thermal proteome profiling Functional Identification of potential drug target and Changes in protein thermal stability in cells can
off-targets result from upstream events (such as changes in
PPIs or PTMs) and may not necessarily indicate
direct drug–protein interaction
It is difficult to ascertain the affinity of the
molecule to its target
PTM profiling Physical, spatial, Elucidation of time- and dose-dependent Enrichment of some PTMs (such as acetylation or
functional and signalling events mediated by PTMs tyrosine phosphorylation) relies on high-affinity
co-regulatory and high-specificity antibodies
High recovery of the modified peptides requires
careful protocol optimization
Large input material requirements can limit the
analysis of tissue and clinical samples
Bottom-up approaches can suffer from ambiguity
in PTM localization assignment
PPI, protein–protein interaction; PTM, post-translational modification.

Nature Reviews | Drug Discovery


Reviews

Box 1 | common methods of quantification in mass spectrometry-based proteomics


Bottom-up proteomics is a commonly used workflow for the mass spectrometry-based characterization of proteins
following proteolytic digestion. Peptides are typically separated by reverse-phase liquid chromatography and introduced
to the mass spectrometer through electrospray ionization of the eluting peptides, where mass-to-charge ratios (m/z)
are measured during the first stage of mass spectrometry (Ms1). in data-dependent acquisition, the most intense intact
precursor ions are selected for fragmentation and detection in Ms2 (see the figure). the Ms2 spectra are typically
matched to peptide sequences through database searching. Label-free quantification, which does not utilize isotope
labelling of the peptides, can be performed on the basis of the Ms1 or Ms2 scans, with the relative quantification coming
from counting peptide spectral matches or assessing signal intensities.
an inherent drawback of bottom-up proteomics is incomplete sequence coverage for many proteins, even with
high-sensitivity protocols and instrumentation. Data-dependent sampling can lead to missing quantitative data between
samples. awareness of this limitation allows for the incorporation of steps to address ambiguity, such as assuring that
distinguishing peptides are quantified when differentiating between protein isoforms128.
stable isotope-based multiplexing approaches can partially overcome missing-data issues by comparing conditions
within a single mass spectrometry run. stable isotope-based approaches include metabolic (such as stable
isotope labelling of amino acids in cell culture (SILAC)67) or chemical (such as, tandem mass tags (tMt)129) labelling.
Metabolic labelling typically allows for analysis of 2 or 3 experimental conditions (such as drug-treated versus untreated
cells), whereas isobaric chemical labelling now allows for comparison of up to 16 different conditions. reporter ions from
isobaric chemical labels can be generated during Ms2 or Ms3. Quantification during Ms3 allows for selective isolation
and fragmentation of the product ions in complex Ms2 mixtures, where fragments from multiple precursor ions with
the same intact m/z may be present.
in targeted quantification, peptides are monitored by mass spectrometry based on a predefined list precursor or
fragment-ion m/z ratios. isotopically labelled internal standards (such aQua peptides130) can improve quantification
when they are introduced during sample processing, especially for low-abundance features (such as phosphopeptides
or isoform-defining peptides). Data-independent acquisition is an increased-throughput-targeted approach, in which
swaths of ions are co-isolated and co-fragmented for concurrent Ms2 analysis131, allowing systematic quantification of
many peptides across samples.
Peptide Precursor ion Product ion
separation dissociation dissociation
Liquid MS1 MS2 MS3
chromatography Intact peptide Peptide fragment Reporter ion
detection detection detection

100 100 100


75 75 75
Intensity

Intensity

50 50 50
25 25 25
0 0 0
400 405 410 415 250 750 1,250 1,750 126 127 128 129 130 131
m/z m/z m/z

Peak area Reporter ion intensity


abundance

abundance
Relative

Relative

Retention 126 127


time (min) m/z

An emerging theme across many studies is that con- inhibition (such as the protein kinase PLK1 inhibitor
served communities of kinases function interchangeably BI-2536 and the JAK2–FLT3 inhibitor fedratinib)61.
to mediate resistance to kinase inhibition. This phenom­ GPCRs are another family of signalling proteins that
enon of adaptive kinome response is common for multitask within numerous protein communities. GPCR
single-agent kinase-targeted therapies and was observed signalling is initiated upon ligand binding and is propa-
during proteomics-based kinome profiling in treatments gated downstream to effector molecules through hetero-
with HER2, MEK and PI3K inhibitors58–60. For instance, trimeric G proteins62. Owing to their essential regulatory
lapatinib-resistant HER2-positive cell lines elicit a functions and accessibility on the cell surface, GPCRs
hetero­geneous adaptive response and show sensitivity are targeted by a wide array of approved and emerging
to inhibition of transcriptional reprogramming medi- therapeutic agents62. Certain GPCR-targeting agents can
ated by bromodomain and extra-terminal domain (BET) differentially impact downstream signal propagation
chromatin readers59. Interestingly, dual agents capable through a mechanism called ‘biased agonism’, which
of targeting both kinases and bromodomain-containing is defined as the property of a molecule to activate one
(BRD) proteins may already exist, with Ciceri et al. signalling pathway while antagonizing another63–65.
reporting on several clinical kinase inhibitors that dis- Selective modulation of GPCR signalling has been
play polypharmacological properties, including BRD4 studied with quantitative proteomics46,66. Christensen et al.

www.nature.com/nrd
Reviews

Thermal-shift assay used stable isotope labelling by amino acids in cell cul- ability of discovery proteomic assays to assess cryptic
A method used to quantify ture (SILAC67) and quantitative phosphoproteomics to protein communities in order to predict and explain the
changes in protein stability analyse the responses elicited by [Sar1,Ile4,Ile8] angio­ unintended side effects of therapeutic agents.
under thermal denaturation in tensin II (SII Ang II), a biased agonist that activates
response to different
treatments or conditions, such
only Gαq-independent signalling, versus Ang II, a full Drugs affect protein localization
as binding of a small molecule. agonist of GPCR Ang II type 1 receptor (AT1R)68. The Many cellular processes are controlled through regulated
study revealed a divergence between Gαq-dependent subcellular localization of key proteins. In one promi-
and Gαq-independent signalling, in which, for example, nent example, the 14-3-3 proteins — which are dedicated
Gαq-mediated phosphorylation of some of the MAPK1/3 to recognizing and sequestering proteins in the cytosolic
substrates was reflective of nuclear translocation down- compartment — control the expression of a broad com-
stream of Ang II, but not of SII Ang II, stimulation68. munity of signalling molecules76. Similar mechanisms
Mass spectrometry proteomics revealed qualitatively regulate localization of the signalling receptor NOTCH77,
and quantitatively different GPCR signalling down- the transcriptional regulators YAP and TAZ78, and the
stream of β2-adrenergic receptor stimulation by two dif- histone-modifying enzyme HDAC4 (ref.79). The recep-
ferential agonists (isoprenaline and salmeterol). Despite tor stability and assembly of membrane-associated
targeting the same signalling nodes, there were differ- signalling complexes is also regulated by PPIs80–82. For
ences in cAMP production, in initial rates of endocytosis the transcriptional regulator and tumour suppressor
and in the phosphorylation of downstream targets, but p53, localization to the mitochondrial outer mem-
not in the late (transcriptional) phase of signal propa- brane may facilitate a novel transcription-independent
gation65. These differences may have important impli- apoptotic programme (reviewed in10). Beyond abun-
cations in the contexts of interindividual differences in dance, proteomic analysis can provide information on
efficacy, unintended activities and resistance to therapy. changes in protein localization upon drug treatment
Because dynamic protein communities shape the (Fig. 1; Table 1).
behaviour of diseased cells and tissues, novel therapeutic Within the proteome, plasma membrane proteins
targets can emerge from proteomic assessment of sig- serve as first-line responders to environmental changes.
nalling nodes, epigenetic regulators and protein degra- Owing to their accessibility, many members of this
dation pathways. With these goals in mind, Bantscheff community also serve as therapeutic targets for
et al. investigated the effects of histone deacetylase small-molecule and antibody-based therapeutics43,45,83.
(HDAC) inhibitors on PPIs using chemoproteomics69. Amongst these, extracellular interactions between
Isolation and quantification of proteins bound to an immune checkpoint receptor programmed cell death
HDAC inhibitor-coupled resin enabled comparisons of protein 1 (PD-1) and its ligand PD-L1 have emerged as
drug potencies, binding kinetics and off-target binding central targets in cancer immunotherapy (reviewed in83).
across the community of HDAC complexes, including Antibody-based therapies that block PD-1 interaction
SIN3, nucleosome remodelling deacetylase (NuRD) with PD-L1 have been tested across many cancer types
and co-repressor to the RE1-silencing transcription and have proved successful for a subset of patients83.
factor (CoREST)69. Separately, the community of pro- A complementary approach to the existing antibody-
teins engaged by the HDAC inhibitor panobinostat based strategy has emerged from proteomic studies
were studied in living cells via thermal proteome profil- of cancer cell lines that were deficient chemokine-
ing through thermal-shift assay70,71. In that work, protein like factor-like MARVEL transmembrane domain-
stability was monitored over a range of temperatures at containing family member 6 (CMTM6), a protein with
a fixed compound concentration — or over a range of previously unknown function80,81. Depletion of CMTM6
compound concentrations at a fixed temperature — in in tumour cells specifically destabilized PD-L1 at the
order to reveal direct and indirect effects of drug binding plasma membrane and restored the cytotoxic activity of
on members of the community71,72. PD-1-expressing T cells80, suggesting a new entry point
The aetiology of off-target effects has been extensively for modulating the immune checkpoint. In the context
explored with ABPP and chemoproteomics (Table 1; of T cell receptor (TCR) activation, a combination of
Fig. 3). The fatty amide hydrolase inhibitor BIA 10-2474 ABPP and proximity labelling showed translocation
displayed off-target interactions with several lipases that of the deubiquitinase USP12 from the nucleus to the
have important metabolic functions in neurons33 and cytosol and stabilization of the TCR complex82, confirm-
may help explain the neurological phenotypes observed ing the utility of mass spectrometry-based approaches
in clinical trials involving this inhibitor73. Similarly, the for shedding light on novel inputs to therapeutically
proteasome inhibitor bortezomib exhibits certain neuro­ meaningful protein communities.
toxic side effects in patients undergoing treatment for
multiple myeloma74. An assessment of serine hydrolase Fractionating subcellular compartments. Even when pro-
activities in this context demonstrated inhibition of tein levels within affected cells are seemingly unchanged,
several additional serine hydrolases by bortezomib74. the spatial distribution of a protein community can be
For the CDK4/6 inhibitors, chemoproteomic profil- dynamic14,37,84. Drug-induced and disease-initiated redis-
ing revealed inhibition of additional protein and lipid tribution of proteins can be studied using different types
kinases by palbociclib in liver cancer cells and primary of microscopy, although reagent availability, multiplexing
tumour tissues, indicating that its polypharmacology and quantification between samples still present chal-
may exert clinical effects not exhibited by the more spe- lenges38. These approaches are complemented by meth-
cific ribociclib75. Together, these studies demonstrate the ods that combine biochemical separation and mass

Nature Reviews | Drug Discovery


Reviews

Synaptic cleft
spectrometry-based quantification, which can provide of biotinylated protein complexes using denaturing
Space or gap at the synapse protein localization on a global scale14,36,37,85,86 (Fig. 3). and non-denaturing conditions enabled the recovery
between a neuron and an The localization of organelle proteins by isotope tagging of labelled cleft proteins and cytoplasmic constituents
effector cell (such as a neuron (LOPIT) approach, developed by Lilley and colleagues, from the PSD12. The identification of community mem-
or a muscle cell).
couples density centrifugation or differential centrifu- bers in these unbounded structures can elucidate basic
Postsynaptic density gation with mass spectrometry to determine the pro- biology and suggest potentially novel therapeutic targets
Protein-dense compartment at tein composition of intracellular organelles36,86. Building in neurological disorders. Looking ahead, a combina-
the postsynaptic membrane of on this technique, Itzhak et al. introduced isotopically tion of proximity labelling with fast kinetics and mass
a synapse between two
labelled organelle fractions as internal references, to spectrometry-based multiplexing has the potential to
neurons.
improve reproducibility and enable the identification resolve the temporal events occurring at neurological
Xenobiotics of protein translocation events between conditions14. synapses.
Substances that are foreign to Protein translocation events associated with EGFR sig- One challenge at the intersection of proteomics and
the biological system. nalling were reported, although these were limited to drug development is the transient nature of the PPIs that
BioID
cell lines. These events included localization of EGFR regulate immediate responses to xenobiotics. Although
A mass spectrometry-based to endosomal compartments upon receptor stimulation they are dynamic and complex, the interaction net-
method for studying the with EGF14, as well as the sequestration of EGFR adaptor works of oncoproteins such as MYC and KRAS have the
associations of a protein of proteins (such as growth factor receptor-bound protein 2 potential to reveal new therapeutic modalities89,90. Both
interest fused to a biotin ligase
(GRB2) and SHC-transforming protein 1 (SHC1)) in the oncoproteins have essential functions in normal physiol­
that labels other proteins in its
close proximity.
cytosol away from the secretory organelles upon treat- ogy and development, and both lack an enzymatic
ment with the EGFR inhibitor gefitinib37. Subcellular activity that can be easily manipulated. These dynamic
fractionation also revealed changes in protein distribu- mechanisms must be thoroughly understood in order
tion that did not manifest as changes in global protein to minimize the side effects associated with therapeutic
levels, including upregulation of mismatch repair pro- targeting. Historically, however, affinity purification
teins in the microsomal and cytosolic fractions and their coupled to mass spectrometry (AP–MS) approaches have
downregulation in the mitochondrial compartment of provided only a static picture of human protein commu-
glucosamine-treated human Hodgkin lymphoma cells84. nities28 (Fig. 3). These approaches have often required the
Organelle fractionation and mass spectrometry optimization of conditions for each target and have been
analysis were recently paired with immunofluorescence susceptible to artificial interactions introduced by cell
microscopy and transcriptomic analysis in order to lysis. A notable example comes from AP–MS studies of
gene­rate a subcellular map of a human cell38. It is yet SCF ubiquitin ligases that have described an exchange
to be demonstrated whether subcellular fractionation of regulatory factors and substrate receptors in a cell lysis
coupled to mass spectrometry can sensitively and repro- buffer within minutes87. To estimate the composition of
ducibly detect protein translocation events between SCF complexes in cells accurately, Reitsma et al. intro-
closely related compartments, such as endosomes and duced a molecular sponge that effectively suppressed
lysosomes — an ability that could be important for both post-lysis exchange by absorbing unbound exchange
large- and small-molecule drug development. factors91. In another example, SILAC-based quantifica-
tion of interaction stability by isotopic differentiation of
Proximity-based labelling. Proximity labelling is interactions as random or targeted (I-DIRT)92 revealed
another way to investigate protein communities, par- fast-exchanging interactions of class I and class II
ticularly those that are hard to isolate using biochem- HDACs93. Proximity-labelling approaches can also
ical approaches because they are not enclosed within capture snapshots of the dynamics within protein com-
a membrane (Fig. 3) . Such communities include a munities in living cells, such as disruption by ibrutinib
synaptic cleft , postsynaptic density (PSD) and other of the complex formed by MYD88, Toll-like receptor 9
non-membrane-bound cellular compartments (for (TLR9) and B cell receptor (BCR) in subtypes of B cell
example, nucleoli in the nucleus and stress granules in lymphoma94. The BioID and AP–MS methods have been
the cytoplasm). In the case of neurological synapses, used in concert to dissect regulatory interactions within
proximity labelling has been informative in several the Hippo pathway95. Low-solubility membrane- and
recent studies12,87. Synaptic proteins were tagged with chromatin-bound proteins such as the transcription fac-
enzymes that catalyse biotinylation of proteins in their tors TEAD1 and TEAD3 were identified in association
close proximity, such as horseradish peroxidase (HRP) with YAP1 by BioID, but not by AP–MS95. In the same
or biotin ligase (BirA), which enabled protein isolation study, Couzens et al. used phosphatase inhibitor (oka-
and identification by mass spectrometry12,87. Uezu et al. daic acid) treatment to test the hypothesis that Hippo
fused proteins that reside specifically in excitatory and pathway interactions are regulated by phosphorylation95.
inhibitory regions of the PSD of mouse brain with BirA, However, slow biotin ligase activity (with signal typically
in order to label the synaptic complexes in the vicinity being accumulated over more than 12 h) made BioID
of the PSD87. Although inhibitory PSD is especially hard incompatible with acute phosphatase inhibitor treatment
to characterize, owing to its thin structure88, proximity (2.5 h). By contrast, phosphorylation-dependent regula-
labelling revealed remarkable complexity87. In a similar tors of the Hippo pathway were successfully identified by
way, the protein composition of synaptic clefts in rat AP–MS95. Together, these studies convincingly demon-
cortical neuron cultures was investigated by Loh et al., strate the applicability of proximity-labelling proteom-
using HRP-conjugated proteins in combination with a ics to studying the mechanisms of drug sensitivity and
membrane-impermeable substrate12. The purification resistance at the level of protein communities.

www.nature.com/nrd
Reviews

Box 2 | How to pick a time point and a drug concentration immediate and delayed responses to drug treatments?
Events that occur within minutes or seconds, such as
appropriate time points and drug concentrations for proteomic analysis of signalling the immediate mechanistic response of an activated
must be defined for each individual experiment. Prior knowledge of the targeted receptor upon engaging its effector molecule, can pro-
pathway or a related process can serve as a reference for selecting initial time points. foundly influence downstream signalling and pathway
For example, receptor tyrosine kinase (rtK)-mediated phosphorylation can be detected
druggability. Is it feasible to track these acute changes
within seconds after stimulation132,133 but is typically profiled within 2–10 min after
ligand binding, and occasionally up to several hours later44,134. similarly, changes in using proteomics?
lysine acetylation135 and ubiquitylation136 levels have been reported within 1 min of The correlation between drug dosage, substrate con-
rtK activation. rtK internalization was observed within 8–20 min after ligand binding, centration and response duration represents a litmus
and its re-emergence on the cell surface was observed after an hour42,44. Continuous test for the efficiency and specificity of target engage-
treatment with tumour necrosis factor (tNF) induces oscillations in the nuclear– ment. When these local events are placed in the con-
cytoplasmic translocation of transcription factor nuclear factor κB (NF-κB) with text of signalling, proteomic tools can help trace signal
a period of 100 min, as well as waves of target gene expression at 1, 3 and 6 h137,138. propagation across a protein community (Fig. 1; Box 2).
these time points seem to be cell-specific, which indicates that it is necessary to By extension, time course and dose–response analy-
optimize time courses on the basis of the model system137,138. ses of phosphorylation dynamics can reveal direct and
several levels of evidence can help determine an appropriate drug concentration for
secondary effectors (Figs 1,3; Table 1). Upon treatment
cell-based experiments. Biochemical IC50 values for a compound and for structurally
related molecules can provide a starting point. small-scale preliminary studies with with lapatinib, dynamic phosphorylation of the down-
western blotting (wB) or immunoprecipitation followed by wB (iP–wB) readouts stream signalling nodes (such as adaptor proteins,
provide evidence on the appropriate time points and drug concentrations in cells transcription factors or cytoskeletal proteins) reflects
prior to large-scale experimentation. although preliminary studies can be cumbersome their physical proximity to the initial EGFR or HER2
— especially for poorly characterized processes in which the reagents require signal104. In melanoma, a time course analysis of phos-
optimization — they can save weeks to months in large-scale proteomic experiments phorylation revealed activation of the DNA damage
by maximizing the chances that the key biological question will be addressed by the response (DDR) pathway upon simultaneous inhibition
experimental design. of RAS–RAF–MEK and PI3K–AKT–mTOR pathways
with cobimetinib (MEK inhibitor) and pictilisib (PI3K
Insights into transient PPIs can reveal novel pathway- inhibitor) and highlighted the opportunity for enhanc-
targeting strategies. The SOX2 transcription factor is ing cytotoxicity through dual pathway inhibition105. In
frequently amplified in squamous cell carcinoma and is diffuse intrinsic pontine glioma, for which limited thera-
itself not readily druggable96. BioID revealed that SOX2 peutics currently exist, strategies have emerged in recent
interacts with the EP300 transcription co-activator, years, such as concurrent inhibition of PI3K–MEK106,
whose inhibition suppressed the growth of lung cancer BET107 and the histone–lysine methyltransferase EZH2
cells96. Split-BioID, which uses the fusion of two inactive (ref.108). Whether individually or in combination, these
halves of BirA to interacting proteins of interest, suc- putative targets affect signalling within multiple protein
cessfully identified transient dimerization-dependent communities and would benefit from systematic pro-
protein interactions of PP1 phosphatase 97 and teomic profiling in preclinical models and beyond.
miRNA-mediated gene-silencing complexes98, and this Beyond identifying key signalling proteins, time-
seems a suitable technique for evaluating compounds resolved analysis of proteins and PTMs can help sub-
that disrupt PPIs. A complementary approach is to fuse divide them into communities that act early, midway
a protein of interest or a small-molecule-interacting through and late in signal propagation. This temporal
protein to the HIV Gag protein99. Gag stimulates the blueprint can establish connections to unappreciated
production of virus-like particles that trap protein com- biological processes and their points of regulation. In
plexes inside themselves, for isolation and subsequent one example, segregation of phosphorylation-mediated
identification by mass spectrometry (Virotrap)99. Proof signalling events was demonstrated following glu-
of concept for the Virotrap approach has been demon- cose stimulation of insulin-secreting beta cells109. The
strated for innate immune-signalling complexes100, phosphopeptide profiles clustered into four distinct
small-molecule binders100 and a RAS-associated ubiq- groups, and the presence of enriched phosphoryl­
uitin ligase substrate adapter, leucine-zipper-like ation motifs among differential responders highlighted
transcriptional regulator 1 (LZTR1)101. the roles of distinct kinases109. Temporal resolution
Recent advances in proteomics have enabled near- of global proteome changes was used to investigate
comprehensive assessment of protein communities antibiotic resistance at sub-lethal doses of rifampicin
that can be isolated biochemically, by affinity purifi- during early exposure, at the onset of bacteriostasis
cation or via proximity labelling (Table 1; Fig. 3). Such and during recovery from treatment110. This analysis
methodologies are now regularly revealing mecha- revealed protein communities that act as the driving
nisms of drug action that would be invisible at the level forces at each stage of antibiotic resistance: an initial SOS
of the global transcriptome or proteome, and they hold response, upregulation of transcription and translation,
promise for otherwise intractable targets96,101–103. and the expression of drug-inactivating enzymes110.
Differential signalling triggered by two EGFR ligands,
Proteomics in space and time EGF and transforming growth factor alpha (TGFα), was
With global proteome-profiling studies revealing resolved by, respectively, interactome, phosphoproteome,
changes in the composition of protein communities, ubiquitinome and proteome analyses across time, so as
questions of temporal regulation arise. What are the pri- to charac­terize the transient and sustained responses
mary and secondary events in signal propagation, or the induced by each ligand44. These signal propagation

Nature Reviews | Drug Discovery


Reviews

Single-shot mass
maps can be superimposed on the community of maturing. For instance, postsynaptic densities isolated
spectrometry proteins regulated by drug-induced changes, in order from mouse brains by immunoaffinity have been charac­
A mass spectrometry to highlight transient regulatory events. terized throughout development119. Following spinal
experimental setup wherein One area where global proteome profiling is set cord injury, soluble factors were quantified across several
single sample injections are
performed, as opposed to
to have a profound impact is in the development of spinal cord segments120. This analysis revealed the inhi-
multiple injections of proteolysis-targeting chimeric (PROTAC) molecules. bition of regenerative and the activation of inflammation
fractionated peptide mixtures. These and other degraders seek to induce the recruit- processes in the first segment posterior to the spinal cord
ment of target substrates to an E3 ligase in order to trig- lesion, presenting a potential new therapeutic target for
ger their ubiquitylation and degradation, most recently stimulating regenerative processes120. In another study,
demonstrated for RTK targeting as an alternative to brain region-specific phosphosignalling maps were
enzymatic inhibition111 (Fig. 1). In this emerging field, obtained for five structurally different GPCR kappa
mass spectrometry-based proteomics is ideally suited to opioid receptor (KOR) agonists121. KOR ligands are major
characterizing on- and off-target effects112. Winter et al. therapeutic candidates for neurological diseases, owing
used proteomics to demonstrate the selectivity of dBET1, to their analgesic effects, but some molecules (for exam-
a tool compound composed of JQ1 and an E3 ubiqui- ple, U-50488H) can produce undesirable effects, such as
tin ligase complex-binding thalidomide moiety that is dysphoria and aversion121. A study revealed activation of
capable of reducing the protein levels of BRD2, BRD3 mTOR signalling by the KOR ligands with undesirable
and BRD4, as well as downregulating gene expression side effects and demonstrated that pre-treatment with
through the MYC oncogene102. The dBET1 molecule mTOR inhibitor can abolish conditioned place aversion
outperformed JQ1 in triggering apoptosis in primary in mice treated with U-50488H121. With the development
AML cells and in reducing tumour growth in a human and accessibility of high-sensitivity mass spectrometry
leukaemia xenograft model102. Similarly, Bondeson et al. instrumentation, such spatiotemporal proteomic studies
demonstrated the specificity of RIPK2- and oestrogen- will hopefully further extend into in vivo drug discovery.
related receptor (ERR) α-targeting degraders in THP-1 Many developmental and clinical targets function at
and MCF-7 cell cultures, respectively103. the intersection of signalling pathways. Characterization
Temporal profiles of the cellular proteome can resolve of the drugs that target these proteins at intersections
changes in drug-induced signalling, but they do not may now be useful for the prediction of off-target
provide information on the spatial changes within and effects and tissue-specific phenotypes. For example, the
between protein communities. Although frequently phospholipase D isoenzymes PLD1 and PLD2 generate
overshadowed by shifts in protein levels, these events can phosphatidic acid and act downstream of many plasma
themselves underlie significant outcomes. One example membrane signalling receptors122. These isoenzymes
is the regulation of PD-L1 levels at the plasma membrane have been studied as targets in cancer, neurodegenera-
by CMTM6 through the inhibition of degradation80,81. tion and infectious disease122. However, the involvement
Proteomic characterization of individual organelles pro- of phosphatidic acid in multiple signalling pathways pre-
vides a ‘zoom-in’ feature for more thorough exploration sents a challenge, owing to toxicity122. The combination
of dynamic protein community maps. of chemoproteomics, proximity labelling and cell line
The localization, PTMs and abundance of proteins engineering (for example, introduction of an ascorbic
can now be resolved across space and time using orga- acid peroxidase (APEX) tag at the endogenous gene
nelle separation or proximity labelling combined with locus) should enable systematic evaluation of com-
multiplexed quantitative methods. For example, prox- pounds targeting phospholipase D isoforms and their
imity labelling with fast kinetics nicely resolved the associated protein communities.
timing of local events in the GPCR community113,114. Cell-to-cell communication in tissues with hetero­
Single-shot mass spectrometry with isobaric labelling geneous cell populations provides another unique
has been used for whole-organelle profiling, as demon- challenge. Proteomic studies of extracellular inter-
strated through the characterization of protein and PTM actions, secreted molecules, and the corresponding
dynamics occurring in isolated mitochondria23,115, and intracellular responses are valuable first steps in dis-
this method holds promise in combination with rapid covering pharma­codynamic biomarkers. Co-culture
organelle isolation116. Protein redistribution across of T cells with antigen-presenting B cells was used to
organelles can also be temporally profiled, as has been study extracellular signalling via proximity-based inter-
demonstrated for human cytomegalovirus infection action tagging123. Tian et al. also used T cell and B cell
(HCMV)117,118. For example, Beltran et al. demonstrated co-culture to characterize CD28 receptor signalling in
the translocation of scavenger receptor class B member 1 the presence of the rheumatoid arthritis drug CTLA4-Ig,
(SCARB1) from lysosomes to the plasma membrane, which can block interaction of CD28 with the B7 ligand
whereas the unconventional myosin MYO18A trafficked on B cells124. Temporal analysis of reciprocal phospho-
from the plasma membrane to the HCMV assembly signalling between stromal and pancreatic tumour
complex at later stages of the infection118. In the future, cells revealed that the signals exchanged between cell
integrating chemoproteomics and ABPP into spatiotem- populations interact in complex ways125. In vivo stud-
poral assessments of protein communities holds pro­ ies of tumour–stroma interactions in cancer cell lines
mise for profiling the effects of drugs on their direct and and patient-derived xenograft models may further
unintended targets. differentiate the tumour proteome from the micro­
Although their development has been challeng- environment, on the basis of human- and mouse-specific
ing, in vivo spatiotemporal proteomic methods are protein sequences126,127. This approach could be used to

www.nature.com/nrd
Reviews

identify the community of proteins that correlates with existing methods towards building a rich understanding
tumour subtype, drug response or acquired resistance. of protein communities in vivo, to embrace and system-
Future efforts will be necessary in order to capitalize on atize automation, to measure the stoichiometric rela-
the available proteomic technologies in the context of tionships between proteins and to improve proteomic
tissues. methods for low-level clinical samples. On the drug
development side, the challenge is to start thinking of
Conclusions connections between phenotypes and protein communi-
Studies from the past decade have demonstrated a ties, rather than just between individual proteins. Doing
continually evolving array of applications for mass so will require using proteomics systematically to under-
spectrometry-based proteomics. These investigations stand the full sequence of events following target engage-
have expanded from exploring the fundamentals of ment and the connections of these events to efficacy,
cellular signalling to aiding in drug discovery and unintended activities and resistance. Our understanding
spatio­temporal profiling in response to drug treatment. of protein communities and their relations has grown
Although much of the available data in the community in large part from proteomic investigations of existing
remain proprietary, we hope that this Review provides therapeutic agents and has set the stage for the next
perspective for those in the proteomics and drug develop- round of impactful contributions to the development
ment communities on the opportunities afforded by the of safe and effective therapeutics.
available technology. Looking ahead, the mass spectro­
metry proteomics community should aim to leverage the Published online xx xx xxxx

1. Milo, R. What is the total number of protein molecules 19. Sharma, K. et al. Ultradeep human phosphoproteome proximal and interacting proteins in mammalian cells.
per cell volume? A call to rethink some published reveals a distinct regulatory nature of Tyr and J. Cell Biol. 196, 801–810 (2012).
values. Bioessays 35, 1050–1055 (2013). Ser/Thr-based signaling. Cell Rep. 8, 1583–1594 36. Dunkley, T. P. J., Watson, R., Griffin, J. L., Dupree, P.
2. Klijn, C. et al. A comprehensive transcriptional portrait (2014). & Lilley, K. S. Localization of organelle proteins by
of human cancer cell lines. Nat. Biotechnol. 33, 20. Choudhary, C. et al. Lysine acetylation targets protein isotope tagging (LOPIT). Mol. Cell. Proteom. 3,
306–312 (2015). complexes and co-regulates major cellular functions. 1128–1134 (2004).
3. Nagaraj, N. et al. Deep proteome and transcriptome Science 325, 834–840 (2009). 37. Orre, L. M. et al. SubCellBarCode: proteome-wide
mapping of a human cancer cell line. Mol. Syst. Biol. 21. Schölz, C. et al. Acetylation site specificities of lysine mapping of protein localization and relocalization.
7, 548 (2011). deacetylase inhibitors in human cells. Nat. Biotechnol. Mol. Cell 73, 166–182 (2019).
4. Wilhelm, M. et al. Mass-spectrometry-based draft 33, 415–423 (2015). This study applies tandem mass tag (TMT)-based
of the human proteome. Nature 509, 582–587 22. Kim, W. et al. Systematic and quantitative assessment quantification for organelle correlation profiling
(2014). of the ubiquitin-modified proteome. Mol. Cell 44, in order to assess changes in protein localization
5. Kim, M.-S. et al. A draft map of the human proteome. 325–340 (2011). upon the inhibition of EGFR signalling in lung
Nature 509, 575–581 (2014). 23. Rose, C. M. et al. Highly multiplexed quantitative cancer cells.
6. Bekker-Jensen, D. B. et al. An optimized shotgun mass spectrometry analysis of ubiquitylomes. 38. Thul, P. J. et al. A subcellular map of the human
strategy for the rapid generation of comprehensive Cell Syst. 3, 395–403 (2016). proteome. Science 356, eaal3321 (2017).
human proteomes. Cell Syst. 4, 587–599 (2017). 24. Hornbeck, P. V. et al. PhosphoSitePlus, 2014: mutations, 39. Gygi, S. P., Rochon, Y., Franza, B. R. & Aebersold, R.
7. Wis´niewski, J. R., Hein, M. Y., Cox, J. & Mann, M. PTMs and recalibrations. Nucleic Acids Res. 43, Correlation between protein and mRNA abundance
A ‘proteomic ruler’ for protein copy number and D512–D520 (2015). in yeast. Mol. Cell. Biol. 19, 1720–1730 (1999).
concentration estimation without spike-in standards. 25. Weerapana, E. et al. Quantitative reactivity profiling 40. Schwanhäusser, B. et al. Global quantification of
Mol. Cell. Proteom. 13, 3497–3506 (2014). predicts functional cysteines in proteomes. Nature mammalian gene expression control. Nature 473,
8. Cooper, H. L., Park, M. H. & Folk, J. E. Posttranslational 468, 790–795 (2010). 337–342 (2011).
formation of hypusine in a single major protein occurs 26. Qu, Z. et al. Proteomic quantification and site-mapping 41. Pettersson, M. & Crews, C. M. PROteolysis TArgeting
generally in growing cells and is associated with of S-nitrosylated proteins using isobaric iodoTMT chimeras (PROTACs) — past, present and future.
activation of lymphocyte growth. Cell 29, 791–797 reagents. J. Proteome Res. 13, 3200–3211 (2014). Drug Discov. Today Technol. 31, 15–27 (2019).
(1982). 27. Erickson, B. K. et al. A strategy to combine sample 42. Kesarwala, A. H., Samrakandi, M. M. &
9. Tan, M. et al. Identification of 67 histone marks and multiplexing with targeted proteomics assays for Piwnica-Worms, D. Proteasome inhibition blocks
histone lysine crotonylation as a new type of histone high-throughput protein signature characterization. ligand-induced dynamic processing and internalization
modification. Cell 146, 1016–1028 (2011). Mol. Cell 65, 361–370 (2017). of epidermal growth factor receptor via altered
10. Green, D. R. & Kroemer, G. Cytoplasmic functions 28. Huttlin, E. L. et al. Architecture of the human receptor ubiquitination and phosphorylation.
of the tumor suppressor p53. Nature 458, 1127 interactome defines protein communities and disease Cancer Res. 69, 976–983 (2009).
(2009). networks. Nature 545, 505–509 (2017). 43. Lemmon, M. A., Schlessinger, J. & Ferguson, K. M.
11. Huangyang, P. & Simon, M. C. Hidden features: 29. Huttlin, E. L. et al. The bioplex network: a systematic The EGFR family: not so prototypical receptor tyrosine
exploring the non-canonical functions of metabolic exploration of the human interactome. Cell 162, kinases. Cold Spring Harb. Perspect. Biol. 6, a020768
enzymes. Dis. Model. Mech. 11, dmm033365 (2018). 425–440 (2015). (2014).
12. Loh, K. H. et al. Proteomic analysis of unbounded 30. Bakalarski, C. E. & Kirkpatrick, D. S. A biologist’s 44. Francavilla, C. et al. Multilayered proteomics reveals
cellular compartments: synaptic clefts. Cell 166, field guide to multiplexed quantitative proteomics. molecular switches dictating ligand-dependent EGFR
1295–1307 (2016). Mol. Cell. Proteom. 15, 1489–1497 (2016). trafficking. Nat. Struct. Mol. Biol. 23, 608–618
This study is the first to use proximity labelling 31. Lapek Jr, J. D. et al. Detection of dysregulated (2016).
to study the synaptic-cleft proteome in neuron protein-association networks by high-throughput 45. Loibl, S. & Gianni, L. HER2-positive breast cancer.
cultures and to report proteins specific to proteomics predicts cancer vulnerabilities. Lancet 389, 2415–2429 (2017).
excitatory versus inhibitory synapses. Nat. Biotechnol. 35, 983–989 (2017). 46. Olsen, J. V. et al. Global, in vivo, and site-specific
13. Zuchero, Y. J. Y. et al. Discovery of novel blood–brain This study uses high-throughput quantitative phosphorylation dynamics in signaling networks.
barrier targets to enhance brain uptake of therapeutic proteomics to define predictors of the sensitivity Cell 127, 635–648 (2006).
antibodies. Neuron 89, 70–82 (2016). of breast cancer cell lines to a panel of 195 drugs, 47. Bose, R. et al. Phosphoproteomic analysis of Her2/neu
14. Itzhak, D. N., Tyanova, S., Cox, J. & Borner, G. H. based on dysregulation of protein–protein signaling and inhibition. Proc. Natl Acad. Sci. USA
Global, quantitative and dynamic mapping of protein interactions. 103, 9773–9778 (2006).
subcellular localization. eLife 5, e16950 (2016). 32. Klaeger, S. et al. The target landscape of clinical 48. Rexer, B. N. et al. Phosphoproteomic mass
15. Aebersold, R. & Mann, M. Mass-spectrometric kinase drugs. Science 358, eaan4368 (2017). spectrometry profiling links Src family kinases to escape
exploration of proteome structure and function. This study reports a comprehensive list of targets from HER2 tyrosine kinase inhibition. Oncogene 30,
Nature 422, 347–355 (2016). for 243 clinical kinase inhibitors through chemical 4163–4174 (2011).
16. Paulo, J. A., Mancias, J. D. & Gygi, S. P. Proteome-wide proteome profiling in four cell lines. 49. Choudhary, C. et al. Mislocalized activation of
protein expression profiling across five pancreatic cell 33. Esbroeck, A. C. M van et al. Activity-based protein oncogenic RTKs switches downstream signaling
lines. Pancreas 46, 690–698 (2017). profiling reveals off-target proteins of the FAAH outcomes. Mol. Cell 36, 326–339 (2009).
17. Giansanti, P., Tsiatsiani, L., Low, T. Y. & Heck, A. J. R. inhibitor BIA 10-2474. Science 356, 1084–1087 50. Guo, A. et al. Signaling networks assembled by
Six alternative proteases for mass spectrometry-based (2017). oncogenic EGFR and c-Met. Proc. Natl Acad. Sci. USA
proteomics beyond trypsin. Nat. Protoc. 11, 34. Rhee, H.-W. et al. Proteomic mapping of mitochondria 105, 692–697 (2008).
993–1006 (2016). in living cells via spatially restricted enzymatic 51. Pan, C., Olsen, J. V., Daub, H. & Mann, M. Global
18. Huttlin, E. L. et al. A tissue-specific atlas of mouse tagging. Science 339, 1328–1331 (2013). effects of kinase inhibitors on signaling networks
protein phosphorylation and expression. Cell 143, 35. Roux, K. J., Kim, D. I., Raida, M. & Burke, B. revealed by quantitative phosphoproteomics.
1174–1189 (2010). A promiscuous biotin ligase fusion protein identifies Mol. Cell. Proteom. 8, 2796–2808 (2009).

Nature Reviews | Drug Discovery


Reviews

52. Zhuang, G. et al. Phosphoproteomic analysis 74. Arastu-Kapur, S. et al. Nonproteasomal targets of the 100. Eyckerman, S. et al. Trapping mammalian protein
implicates the mTORC2–FoxO1 axis in VEGF signaling proteasome inhibitors bortezomib and carfilzomib: complexes in viral particles. Nat. Commun. 7, 11416
and feedback activation of receptor tyrosine kinases. a link to clinical adverse events. Clin. Cancer Res. 17, (2016).
Sci. Signal. 6, ra25 (2013). 2734–2743 (2011). This study describes a novel approach for
53. Yamaguchi, H., Chang, S.-S., Hsu, J. L. & Hung, M.-C. 75. Sumi, N. J., Kuenzi, B. M., Knezevic, C. E., Rix, L. L. R. identifying membrane–protein interactions and
Signaling cross-talk in the resistance to HER family & Rix, U. Chemoproteomics reveals novel protein and small-molecule binders by trapping them within
receptor targeted therapy. Oncogene 33, 1073–1081 lipid kinase targets of clinical CDK4/6 inhibitors in viral-like particles (Virotrap).
(2014). lung cancer. ACS Chem. Biol. 10, 2680–2686 (2015). 101. Steklov, M. et al. Mutations in LZTR1 drive human
54. Boyer, A. P., Collier, T. S., Vidavsky, I. & Bose, R. 76. Muslin, A. J., Tanner, J. W., Allen, P. M. & Shaw, A. S. disease by dysregulating RAS ubiquitination. Science
Quantitative proteomics with siRNA screening Interaction of 14–3–3 with signaling proteins is 362, 1177–1182 (2018).
identifies novel mechanisms of trastuzumab resistance mediated by the recognition of phosphoserine. 102. Winter, G. E. et al. Phthalimide conjugation as a
in HER2 amplified breast cancers. Mol. Cell. Proteom. Cell 84, 889–897 (1996). strategy for in vivo target protein degradation. Science
12, 180–193 (2013). 77. Ramakrishnan, G. et al. AKT and 14-3-3 regulate 348, 1376–1381 (2015).
55. Liu, L. et al. Novel mechanism of lapatinib resistance Notch4 nuclear localization. Sci. Rep. 5, 8782 (2015). 103. Bondeson, D. P. et al. Catalytic in vivo protein
in HER2-positive breast tumor cells: activation of AXL. 78. Kanai, F. et al. TAZ: a novel transcriptional co-activator knockdown by small-molecule PROTACs. Nat. Chem.
Cancer Res. 69, 6871–6878 (2009). regulated by interactions with 14-3-3 and PDZ Biol. 11, 611–617 (2015).
56. Liu, W. et al. Quantitative proteomics profiling reveals domain proteins. EMBO J. 19, 6778–6791 (2000). 104. Imami, K. et al. Temporal profiling of lapatinib-
activation of mTOR pathway in trastuzumab resistance. 79. Wang, A. H. et al. Regulation of histone deacetylase 4 suppressed phosphorylation signals in EGFR/HER2
Oncotarget 8, 45793–45806 (2017). by binding of 14-3-3 proteins. Mol. Cell Biol. 20, pathways. Mol. Cell. Proteom. 11, 1741–1757
57. Li, J. et al. A chemical and phosphoproteomic 6904–6912 (2000). (2012).
characterization of dasatinib action in lung cancer. 80. Mezzadra, R. et al. Identification of CMTM6 and 105. Kirkpatrick, D. S. et al. Phosphoproteomic
Nat. Chem. Biol. 6, 291–299 (2010). CMTM4 as PD-L1 protein regulators. Nature 549, characterization of DNA damage response in
58. Duncan, J. S. et al. Dynamic reprogramming of the 106–110 (2017). melanoma cells following MEK/PI3K dual inhibition.
kinome in response to targeted MEK inhibition in 81. Burr, M. L. et al. CMTM6 maintains the expression Proc. Natl Acad. Sci. USA 110, 19426–19431
triple negative breast cancer. Cell 149, 307–321 of PD-L1 and regulates anti-tumour immunity. Nature (2013).
(2012). 549, 101–105 (2017). 106. Wu, Y. L. et al. Dual inhibition of PI3K/AKT and
This study uses multiplexed kinase inhibitor 82. Jahan, A. S. et al. Usp12 stabilizes the T-cell receptor MEK/ERK pathways induces synergistic antitumor
beads and mass spectrometry (MIB-MS) to define complex at the cell surface during signaling. Proc. Natl effects in diffuse intrinsic pontine glioma cells. Transl
kinome reprogramming in triple-negative breast Acad. Sci. USA 113, E705–E714 (2016). Oncol. 10, 221–228 (2017).
cancer cell lines treated with the MEK inhibitor 83. Kim, J. M. & Chen, D. S. Immune escape to PD-L1/PD-1 107. Piunti, A. et al. Therapeutic targeting of polycomb and
AZD6244. blockade: seven steps to success (or failure). Ann. Oncol. BET bromodomain proteins in diffuse intrinsic pontine
59. Stuhlmiller, T. J. et al. Inhibition of lapatinib-induced 27, 1492–1504 (2016). gliomas. Nat. Med. 23, 493–500 (2017).
kinome reprogramming in ERBB2-positive breast 84. Carvalho, A. S., Molina, H. & Matthiesen, R. New 108. Mohammad, F. et al. EZH2 is a potential therapeutic
cancer by targeting BET family bromodomains. insights into functional regulation in MS-based drug target for H3K27M-mutant pediatric gliomas.
Cell Rep. 11, 390–404 (2015). profiling. Sci. Rep. 6, srep18826 (2016). Nat. Med. 23, 483–492 (2017).
60. McNeill, R. S. et al. Combination therapy with potent 85. Foster, L. J. et al. A mammalian organelle map by 109. Sacco, F. et al. Glucose-regulated and drug-perturbed
PI3K and MAPK inhibitors overcomes adaptive protein correlation profiling. Cell 125, 187–199 phosphoproteome reveals molecular mechanisms
kinome resistance to single agents in preclinical (2006). controlling insulin secretion. Nat. Commun. 7, 13250
models of glioblastoma. Neuro-Oncol. 19, 86. Geladaki, A. et al. Combining LOPIT with differential (2016).
1469–1480 (2017). ultracentrifugation for high-resolution spatial 110. Giddey, A. D. et al. A temporal proteome dynamics
61. Ciceri, P. et al. Dual kinase-bromodomain inhibitors proteomics. Nat. Commun. 10, 331 (2019). study reveals the molecular basis of induced
for rationally designed polypharmacology. Nat. Chem. 87. Uezu, A. et al. Identification of an elaborate complex phenotypic resistance in mycobacterium smegmatis
Biol. 10, 305–312 (2014). mediating postsynaptic inhibition. Science 353, at sub-lethal rifampicin concentrations. Sci. Rep. 73,
62. Lappano, R. & Maggiolini, M. G protein-coupled 1123–1129 (2016). 43858 (2017).
receptors: novel targets for drug discovery in cancer. 88. Tao, C.-L. et al. Differentiation and characterization 111. Burslem, G. M. et al. The advantages of targeted
Nat. Rev. Drug. Discov. 10, 47–60 (2011). of excitatory and inhibitory synapses by cryo-electron protein degradation over inhibition: an RTK case
63. Thompson, G. L., Kelly, E., Christopoulos, A. & tomography and correlative microscopy. J. Neurosci. study. Cell Chem. Biol. 25, 67–77.e3 (2018).
Canals, M. Novel GPCR paradigms at the μ-opioid 38, 1493–1510 (2018). 112. Sakamoto, K. M. et al. Development of protacs to
receptor. Br. J. Pharmacol. 172, 287–296 (2015). 89. Kalkat, M. et al. MYC protein interactome profiling target cancer-promoting proteins for ubiquitination
64. Gundry, J., Glenn, R., Alagesan, P. & Rajagopal, S. reveals functionally distinct regions that cooperate and degradation. Mol. Cell. Proteom. 2, 1350–1358
A practical guide to approaching biased agonism at to drive tumorigenesis. Mol. Cell 72, 836–848.e7 (2003).
G protein coupled receptors. Front. Neurosci. 11, 17 (2018). 113. Paek, J. et al. Multidimensional tracking of GPCR
(2017). 90. Adhikari, H. & Counter, C. M. Interrogating the protein signaling via peroxidase-catalyzed proximity labeling.
65. Tsvetanova, N. G. et al. G protein–coupled receptor interactomes of RAS isoforms identifies PIP5K1A as Cell 169, 338–349.e11 (2017).
endocytosis confers uniformity in responses to a KRAS-specific vulnerability. Nat. Commun. 9, 3646 114. Lobingier, B. T. et al. An approach to spatiotemporally
chemically distinct ligands. Mol. Pharmacol. 91, (2018). resolve protein interaction networks in living cells.
145–156 (2017). 91. Reitsma, J. M. et al. Composition and regulation Cell 169, 350–360 (2017).
66. Williams, G. R. et al. Exploring G protein-coupled of the cellular repertoire of SCF ubiquitin ligases. 115. Ordureau, A. et al. Dynamics of PARKIN-dependent
receptor signaling networks using SILAC-based Cell 171, 1326–1339 (2017). mitochondrial ubiquitylation in induced neurons
phosphoproteomics. Methods 92, 36–50 (2016). 92. Tackett, A. J. et al. I-DIRT, a general method for and model systems revealed by digital snapshot
67. Ong, S.-E. Stable isotope labeling by amino acids in distinguishing between specific and nonspecific proteomics. Mol. Cell 70, 211–227 (2018).
cell culture, SILAC, as a simple and accurate approach protein interactions. J. Proteome Res. 4, 1752–1756 116. Chen, W. W., Freinkman, E., Wang, T., Birsoy, K.
to expression proteomics. Mol. Cell. Proteom. 1, (2005). & Sabatini, D. M. Absolute quantification of matrix
376–386 (2002). 93. Joshi, P. et al. The functional interactome landscape of metabolites reveals the dynamics of mitochondrial
68. Christensen, G. L. et al. Quantitative phosphoproteomics the human histone deacetylase family. Mol. Syst. Biol. metabolism. Cell 166, 1324–1337 (2016).
dissection of seven-transmembrane receptor signaling 9, 672 (2013). 117. Weekes, M. P. et al. Quantitative temporal viromics:
using full and biased agonists. Mol. Cell. Proteom. 9, 94. Phelan, J. D. et al. A multiprotein supercomplex an approach to investigate host–pathogen interaction.
1540–1553 (2010). controlling oncogenic signaling in lymphoma. Nature Cell 157, 1460–1472 (2014).
69. Bantscheff, M. et al. Chemoproteomics profiling of 560, 387–391 (2018). 118. Beltran, P. M., Mathias, R. A. & Cristea, I. M.
HDAC inhibitors reveals selective targeting of HDAC This study uses SILAC-based quantification to A portrait of the human organelle proteome in space
complexes. Nat. Biotechnol. 29, 255–265 (2011). characterize a multiprotein subcomplex regulating and time during cytomegalovirus infection. Cell Syst.
70. Molina, D. M. et al. Monitoring drug target BCR signalling in ibrutinib-responsive diffuse large 3, 361–373 (2016).
engagement in cells and tissues using the cellular B cell lymphoma. 119. Li, J. et al. Spatiotemporal profile of postsynaptic
thermal shift assay. Science 341, 84–87 (2013). 95. Couzens, A. et al. Protein interaction network of the interactomes integrates components of complex brain
This seminal study describes the development and mammalian hippo pathway reveals mechanisms of disorders. Nat. Neurosci. 20, 1150–1161 (2017).
application of cellular thermal shift assay (CETSA) kinase-phosphatase interactions. Sci. Signal. 6, rs15 120. Devaux, S. et al. Proteomic analysis of the spatio-
for evaluating target engagement by a drug in (2013). temporal based molecular kinetics of acute spinal cord
intact cells. 96. Kim, B. R. et al. Identification of the SOX2 interactome injury identifies a time- and segment-specific window
71. Franken, H. et al. Thermal proteome profiling for by BioID reveals EP300 as a mediator of SOX2- for effective tissue repair. Mol. Cell. Proteom. 15,
unbiased identification of direct and indirect drug dependent squamous differentiation and lung 2641–2670 (2016).
targets using multiplexed quantitative mass squamous cell carcinoma growth. Mol. Cell. Proteom. 121. Liu, J. J. et al. In vivo brain GPCR signaling elucidated
spectrometry. Nat. Protoc. 10, 1567–1593 (2015). 16, 1864–1888 (2017). by phosphoproteomics. Science 360, eaao4927
72. Savitski, M. M. et al. Tracking cancer drugs in living 97. De Munter, S. et al. Split-BioID: a proximity (2018).
cells by thermal profiling of the proteome. Science biotinylation assay for dimerization-dependent protein 122. Brown, H. A., Thomas, P. G. & Lindsley, C. W.
346, 1255784 (2014). interactions. FEBS Lett. 591, 415–424 (2017). Targeting phospholipase D in cancer, infection and
This study builds on the CETSA assay by 98. Schopp, I. M. et al. Split-BioID a conditional neurodegenerative disorders. Nat. Rev. Drug Discov.
incorporating TMT and quantitative mass proteomics approach to monitor the composition 16, 351–367 (2017).
spectrometry for proteome-wide analysis of of spatiotemporally defined protein complexes. 123. Liu, Q. et al. A proximity-tagging system to identify
on- and off-target drug effects. Nat. Commun. 8, 15690 (2017). membrane protein–protein interactions. Nat. Methods
73. Kerbrat, A. et al. Acute neurologic disorder from 99. Titeca, K. et al. Analyzing trapped protein complexes 15, 715–722 (2018).
an inhibitor of fatty acid amide hydrolase. N. Engl. by Virotrap and SFINX. Nat. Protoc. 12, 881–898 124. Tian, R. et al. Combinatorial proteomic analysis of
J. Med. 375, 1717–1725 (2016). (2017). intercellular signaling applied to the CD28 T-cell

www.nature.com/nrd
Reviews

costimulatory receptor. Proc. Natl Acad. Sci. USA 112, 131. Gillet, L. C. et al. Targeted data extraction of the 137. Nelson, D. E. et al. Oscillations in NF-κB signaling
E1594–E1603 (2015). MS/MS spectra generated by data-independent control the dynamics of gene expression. Science 306,
125. Tape, C. J. et al. Oncogenic KRAS regulates tumor acquisition: a new concept for consistent and 704–708 (2004).
cell signaling via stromal reciprocation. Cell 165, accurate proteome analysis. Mol. Cell. Proteom. 11, 138. Tian, B., Nowak, D. E. & Brasier, A. R. A TNF-induced
910–920 (2016). O111.016717 (2012). gene expression program under oscillatory NF-κB
126. Naba, A., Clauser, K. R., Lamar, J. M., Carr, S. A. & 132. Zheng, Y. et al. Temporal regulation of EGF signalling control. BMC Genomics 6, 137 (2005).
Hynes, R. O. Extracellular matrix signatures of human networks by the scaffold protein Shc1. Nature 499,
mammary carcinoma identify novel metastasis 166–171 (2013). Acknowledgements
promoters. eLife 3, e01308 (2014). 133. Reddy, R. J. et al. Early signaling dynamics of the The authors thank members of the MPL group for their input
127. Wang, X. et al. Breast tumors educate the proteome epidermal growth factor receptor. Proc. Natl Acad. and support.
of stromal tissue in an individualized but coordinated Sci. USA 113, 3114–3119 (2016).
manner. Sci. Signal. 10, eaam8065 (2017). 134. Dürnberger, G. et al. Global analysis of muscle-specific Author contributions
128. Bamberger, C. et al. Deducing the presence of kinase signaling by quantitative phosphoproteomics. The authors contributed equally to all aspects of the article.
proteins and proteoforms in quantitative proteomics. Mol. Cell. Proteom. 13, 1993–2003 (2014).
Nat. Commun. 9, 2320 (2018). 135. Bryson, B. D. & White, F. M. Quantitative profiling of Competing interests
129. Thompson, A. et al. Tandem mass tags: a novel lysine acetylation reveals dynamic crosstalk between H.G.B. and D.S.K. are employees of Genentech Inc., a wholly
quantification strategy for comparative analysis of receptor tyrosine kinases and lysine acetylation. owned subsidiary of Roche. D.S.K. is a shareholder in Roche.
complex protein mixtures by MS/MS. Anal. Chem. 75, PLOS ONE 10, e0126242 (2015).
1895–1904 (2003). 136. Akimov, V., G. Rigbolt, K. T., M. Nielsen, M. & Publisher’s note
130. Kirkpatrick, D. S., Gerber, S. A. & Gygi, S. P. The Blagoev, B. Characterization of ubiquitination Springer Nature remains neutral with regard to jurisdictional
absolute quantification strategy: a general procedure dependent dynamics in growth factor receptor claims in published maps and institutional affiliations.
for the quantification of proteins and post-translational signaling by quantitative proteomics. Mol. Biosyst.
modifications. Methods 35, 265–273 (2005). 7, 3223–3233 (2011). © Springer Nature Limited 2020

Nature Reviews | Drug Discovery

You might also like