Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2022 Feb 14;151(1):7–19. doi: 10.1002/ijc.33954

Mapping oncogenic protein interactions for precision medicine

Mehdi Sharifi Tabar 1,2,3, Habib Francis 1,2,3, Dannel Yeo 3,4,5, Charles G Bailey 1,2,3,, John E J Rasko 1,3,4,5,
PMCID: PMC9306658  PMID: 35113472

Abstract

Normal protein‐protein interactions (normPPIs) occur with high fidelity to regulate almost every physiological process. In cancer, this highly organised and precisely regulated network is disrupted, hijacked or reprogrammed resulting in oncogenic protein‐protein interactions (oncoPPIs). OncoPPIs, which can result from genomic alterations, are a hallmark of many types of cancers. Recent technological advances in the field of mass spectrometry (MS)‐based interactomics, structural biology and drug discovery have prompted scientists to identify and characterise oncoPPIs. Disruption of oncoPPI interfaces has become a major focus of drug discovery programs and has resulted in the use of PPI‐specific drugs clinically. However, due to several technical hurdles, studies to build a reference oncoPPI map for various cancer types have not been undertaken. Therefore, there is an urgent need for experimental workflows to overcome the existing challenges in studying oncoPPIs in various cancers and to build comprehensive reference maps. Here, we discuss the important hurdles for characterising oncoPPIs and propose a three‐phase multidisciplinary workflow to identify and characterise oncoPPIs. Systematic identification of cancer‐type‐specific oncogenic interactions will spur new opportunities for PPI‐focused drug discovery projects and precision medicine.

Keywords: cancer, drug discovery, genomic alteration, precision medicine, protein‐protein interaction


graphic file with name IJC-151-7-g002.jpg


Abbreviations

AP‐MS

affinity purification mass spectrometry

APEX

ascorbate peroxidase

BioID

proximity‐dependent biotin identification

cryo‐EM

cryogenic electron microscopy

DLS

dynamic light scattering

FBDD

fragment‐based drug discovery

FRET

Förster resonance energy transfer

HTS

high‐throughput screening

NMR

nuclear magnetic resonance

normPPI

normal protein‐protein interaction

oncoPPI

oncogenic protein‐protein interaction

PBD

peptide binding domain

PL‐MS

proximity labelling mass spectrometry

PPI

protein‐protein interaction

SEC‐MALLS

size exclusion chromatography—multiangle laser light scattering

TAD

topologically associating domain

XL‐MS

cross‐linking mass spectrometry

Y2H

yeast two‐hybrid

1. INTRODUCTION

Multimodal combinations of radiotherapy, surgery, biologicals and chemotherapy have become the mainstay of cancer therapeutics. 1 , 2 These modalities often lack selectivity which can lead to adverse effects such as chemotherapy‐induced cardiotoxicity. 3 , 4 Quality of life in surviving patients is thereby compromised. In contrast, targeted molecular therapies offer a great potential to combat various cancer types with minimal harm to normal cells or organs. 5 , 6 , 7 The main purpose of molecular therapy in oncology is to target specific biological pathways activated by oncoproteins in tumour cells. Oncoproteins are involved in various signalling pathways and mediate a large number of oncogenic protein‐protein interactions (oncoPPIs). 8 , 9 , 10 , 11 OncoPPI interfaces offer great potential for rational drug development and targeted cancer therapy. 8 , 9 , 12 , 13 , 14 , 15 , 16 , 17 , 18 , 19

Recent technological advances in the fields of drug discovery and structural biology have accelerated the characterisation of oncogenic protein interfaces and rational drug design. 20 , 21 In recent years, several oncoPPI modulators have been tested in the clinical setting, with some now approved for clinical use. 17 , 22 , 23 However, most PPI‐based drug discovery projects have focused on a relatively limited number of oncoPPIs detected in focused small‐scale studies. Therefore, identification of more cancer‐specific oncoPPIs in a proteome‐wide manner will increase the potential for drug target discovery, improving options for targeted therapies.

Cancer is a highly complex disease featuring complex genotypes and gene dysregulation, which can result in the formation of complex oncogenic interaction networks. 19 , 24 , 25 , 26 Better understanding of the composition of protein interaction networks of each cancer will greatly enhance our molecular understanding of cancer biology and provide us with the ability to improve patient outcomes through precision and personalised medicine. 11 , 27 Efforts to map oncoPPI networks have already been initiated, for example, lung cancer‐specific oncoPPI networks (~400 PPIs) have been generated on a small‐scale for a few well‐known cancer genes. 11

Several publicly available databases such as BioGrid, 28 BioPlex, 29 IntAct, 30 MINT 31 and HIPPIES 32 regularly update human protein interaction networks by curating PPIs from both focused low‐throughput experimental studies and large high‐throughput mass spectrometry (MS) and yeast two‐hybrid (Y2H) datasets. Databases such as PINA 33 integrate cancer‐focused transcriptomics and proteomics information with PPI datasets to draw a cancer‐specific protein interaction network. However, these maps do not distinguish normal PPIs (normPPIs) from oncoPPIs because nearly all affinity purification MS (AP‐MS)‐generated PPI datasets have been constructed using transformed cell lines, which contain abnormal karyotypes and thus an abnormal interactome profile. Hence, a focus on generating comprehensive maps of the oncoPPI networks for human cancers is crucial.

The protein interaction networks of oncoproteins which are referred to as “oncoPPIs” first appeared in the literature in 1997. 34 Since then, these terms have been used in many publications mostly for well‐known oncoproteins (eg, KRAS) and tumour suppressors (eg, P53), which are categorised as cancer genes in many cancers. 35 Recent large‐scale genome‐wide analysis of 33 different cancer types involving 10 000 tumour samples reported that 55% (ie, 142 out of 258) of cancer genes and their mutations are associated with a single cancer. 35 This strongly suggests that oncoPPI networks could be unique for many cancer types as their cancer driver genes and genomic alterations vary. Here, we define oncoPPIs as any direct PPIs that arise from genomic alterations in tumour cells. Identification and characterisation of oncoPPIs for each cancer can guide targeted molecular therapies and precision medicine. In the following sections, we discuss the genomic alterations and approaches to build reference oncoPPI maps for various cancers.

2. GENOMIC ALTERATIONS RESULT IN COMPLEX oncoPPI NETWORKS

The genomic landscapes of many tumours from diverse cancers have been described. Coding and noncoding mutations, as well as structural variants such as copy number variations, rearrangements, insertions, inversions and translocations have each been defined and characterised. 35 , 36 , 37 , 38 , 39 How pathogenic genomic alterations lead to complex oncogenic traits has remained one of the major challenges in cancer biology. Part of the answer lies in defining how these alterations are linked to oncogenic protein networks, thus enabling a network level understanding of the effects of genomic alterations on cellular functions. 40 , 41 , 42 , 43

The majority of oncoPPIs arise from genomic alterations that directly affect protein interactions via different molecular mechanisms, including edgetic perturbation and/or node removal 42 (Figure 1A). In edgetic perturbation, mutations can change the folding free energy of interacting partners or perturb covalent interactions 43 , 44 and involves change of function. In node removal, genomic alterations lead to the aggregation or misfolding of the target protein, thereby causing the loss of its entire interaction network. 9 Nodal removal involves loss of function. Genomic alteration in regulatory regions can also indirectly affect protein interactions by dysregulating the expression or mislocalisation of target proteins.

FIGURE 1.

FIGURE 1

Genomic alterations, gene dysregulation and protein mislocalisation in cancer give rise to oncoPPIs. (A) Schematic illustration of edgetic perturbation and node removal in cancer and their impact on normal protein interaction networks. Missense mutations are depicted as coloured stars. Specific interactions can be interrupted (dotted line, blue star) or enhanced (thick line, brown star) or created (green star) by a missense mutation, whereas complete loss of the node protein (dotted circle) leads to the loss of the entire interaction network. (B) Gene dysregulation is another source of oncogenic network formation. In normal cells (upper panel), Gene 1 (ie, oncogene) is inactive whereas Gene 2 (ie, tumour suppressor) is active and normPPIs occur. In cancer (lower panel), Gene 1 is activated and initiates oncoPPIs whilst Gene 2 is silenced and normPPIs are lost. (C) Genome organising proteins such as CTCF regulate genome folding globally in a highly organised manner. In cancer, CTCF binding sites or CTCF itself can be mutated leading to loss of CTCF occupancy. This causes disorganisation of genome architecture and dysregulation of neighbouring genes. Dysregulated genes can potentially define new interaction networks. (D) In cancer, mislocalisation of proteins can also initiate new interaction networks. For example, MUC1 and EGFR are normally expressed separately at the cell surface. However, in cancer, MUC1 forms oncogenic PPIs with EGFR, regulating its nuclear localisation and hence transcriptional regulation of target genes

2.1. Genomic alterations can directly affect PPI networks

Analysis of pan‐cancer genome studies show that noncoding mutations are less frequent than coding mutations. About 91% of tumours carry at least one driver mutation, with an average of 2.6 coding mutations per tumour. 36 , 37 The number of large‐scale studies reporting the effects of coding mutations on disease‐specific PPI networks has increased rapidly. 40 , 41 A recent comprehensive investigation of over 10 000 tumour exomes demonstrated that disease‐associated germline variants as well as somatic missense mutations are significantly enriched in PPI interfaces compared to germline variants found in healthy individuals. 40 Further characterisation of some of the variants confirmed that disease‐associated variants disrupt PPIs and regulate tumour growth. 40 In a large‐scale study using the binary Y2H system, PPIs were assessed for ~2500 pathogenic missense mutations. 41 Approximately, 70% of the missense mutations had no effect on protein stability and folding, however half of these mutations resulted in edgetic perturbation. In a comprehensive assessment of somatic missense mutations within 304 cancer‐specific PPI interfaces for which structural data was available, 16 interfaces (~5%) were disrupted. 45

Missense or truncation mutations affecting protein interactions can rewire biological processes in cancer as well as developmental disorders. 41 , 46 This has been observed in melanoma where two missense mutations in cyclin‐dependent kinase 4 (CDK4), R24H and R24C, have been shown to confer resistance to cyclin‐dependent kinase inhibitor 2A (CDKN2A) inhibitors. 47 In a pairwise interaction study, it was shown that CDK4 R24C and R24H, but not other putative disease‐causing mutations (N41S and S52N), disrupted CDK4 binding to CDKN2C. 48 Notably, perturbation of the CDK4‐CDKN2C complex leads to drug resistance phenotypes in melanoma patients.

In a recent report in hereditary pancreatic cancer, it was shown that a truncated RAB‐like protein 3 (RABL3) mutant markedly increased its binding affinity to RAP1 GTPase‐GDP dissociation stimulator 1 (RAP1GDS1) protein, a chaperon involved in prenylation of RAS GTPases. Consequently, the RAS signalling pathway was disrupted leading to enhanced cell proliferation and tumour growth. 49 In another study, it was shown that a missense mutation, R183W, in serine/threonine‐protein phosphatase 2A regulatory subunit 1A (PPP2R1A) disrupted its binding to the canonical subunits of the PP2A complex B module, including B55 and B56 proteins, and abrogated growth suppressive effects. 50 However, mutant PPP2R1A also exhibited increased binding to striatin subunit family members, demonstrating that a missense mutation can confer both loss‐ and gain‐of‐function phenotypes. 50 , 51

2.2. Genomic alterations modulate PPI networks via gene dysregulation

Many cancers from diverse tissues exhibit dysregulated gene expression despite no significant genomic alterations in the protein coding region of the differentially expressed genes. Single nucleotide variants within cis‐regulatory regions of cancer genes have been shown to greatly modulate gene expression in a range of cancers. 52 , 53 These mutations can lead to transcriptional activation or repression of the target genes. For example, the promoter region of telomerase reverse transcriptase (TERT) is frequently mutated in a range of cancers 54 , 55 , 56 and linked to increased tumour growth and metastasis (Figure 1B). Genomic alterations can also occur in trans‐regulatory elements, distant from cis‐regulatory elements of the given cancer genes. For instance, mutations or structural variations in DNA motifs bound by genome‐organising proteins, such as CCCTC‐binding factor (CTCF) and cohesin, or transcription factors, indirectly lead to the dysregulation of many genes in various cancers 57 , 58 , 59 (Figure 1C). Mutations that occur within topologically associating domain (TAD) boundaries or in enhancer and insulator regions can result in coordinated dysregulation of a cluster of neighbouring genes located within the same TAD. 57 , 58 , 59 , 60 Mutations in CTCF itself, and other genome organising proteins such as the cohesin subunit STAG2, can have a similar impact. 61 , 62

In gastrointestinal cancer, a CTCF‐binding motif on chromosome 6 is disrupted by mutations at TAD boundaries flanking centromere protein Q (CENPQ) and methylmalonyl‐CoA mutase (MUT) genes 57 (Figure 1C). In acute myeloid leukaemia, the oncogenic MDS1 and EVI1 Complex Locus (MECOM) protein is activated due to a genomic inversion event that occurs in neighbouring TADs which disrupts CTCF‐mediated genome organisation. 63 Such aberrant activation of proteins can potentiate illicit oncoPPIs and redefine the oncogenic potential of cancer cells. 64 As another example, the aberrant expression of CTCF‐like (CTCFL), a paralogue of CTCF, 65 in treatment‐resistant neuroblastoma alters genome folding and leads to the formation of superenhancers that consequently activate proneural proteins and promote a resistance phenotype. 66 Another example is the bromodomain (BRD)‐containing protein family members, which are readers of lysine acetylation, that are highly elevated in many cancers. BRD4 overexpression leads to the formation of an oncogenic complex with positive transcription elongation factor (p‐TEFb) leading to activation of RNA Polymerase II (RNAPII) at late mitosis. 67

Together, these examples highlight the impact that acquired mutations and structural variations can have on oncoPPI networks. These can directly rewire new oncoPPI networks or indirectly lead to the dysregulation of many genes which then form aberrant protein interaction networks to promote tumour growth and metastasis. Therefore, defining oncoPPIs for each cancer not only sheds light on molecular mechanisms of malignant phenotypes, but also paves the way to design more effective treatments.

2.3. Mislocalisation of oncoproteins can initiate new classes of oncoPPI networks

Spatial compartmentalisation of proteins and organelle‐specific interactions enable cells to conduct numerous biological processes in parallel. However, the subcellular relocalisation of the proteins as a rapid cellular response to internal or environmental stimuli can elicit new protein interaction networks. An exemplar is the receptor tyrosine‐protein kinase HER2, an oncogene associated with progression of aggressive types of breast cancer. HER2 is expressed on the cell surface where it can function as a receptor or can localise to the nucleus upon stimulation to modulate transcription of target genes. 68 , 69 Another example is alpha‐enolase (ENO1), a potential prognostic marker which is elevated in various types of cancers and is linked to tumourigenesis. 70 ENO1 can exhibit distinct functions in different cellular locations: (a) as a gene transcriptional repressor in the nucleus, where it exhibits enzymatic function in the cytosol regulating the phosphatidylinositol 3‐kinase (PI3K)/protein kinase B (AKT) signalling pathway; and (b) as a plasminogen receptor in the plasma membrane. 71 , 72 Similarly, mucin‐1 (MUC1), a well‐characterised oncoprotein extensively expressed on the surface of cancer cells, is also expressed in the nucleus, cytosol and mitochondria, and has been shown to interact with different protein partners in a location‐dependent manner. 73 , 74 , 75 When MUC1 is localised to the nucleus in breast cancer cells it interacts with epidermal growth factor receptor (EGFR), facilitating chromatin binding and thereby rewiring oncogenic pathways 76 (Figure 1D). Therefore, specific biochemical and experimental strategies need to be designed to investigate multilocalising protein interactome networks as they can interact with a range of protein classes as they shuttle between different compartments.

3. EXISTING CHALLENGES FOR ONCOGENIC PROTEIN INTERACTION STUDIES

Genomics, 36 , 37 transcriptomics 45 and proteomics 77 , 78 initiatives coupled with bioinformatics and biostatistics have extracted vast collections of biological data from comparatively small amount of patient material obtained at biopsy. In contrast, system‐wide interactome studies using MS‐based approaches (eg, cross‐linking MS [XL‐MS]) require a large amount of input materials (eg, microgram to milligramme protein sample in a native or seminative condition), which is nearly impossible to extract from limited patient samples. 79 , 80 In addition, AP‐MS and proximity labelling MS (PL‐MS) require gene transfer and genetic manipulation, which is not feasible in patient samples. 81 , 82 , 83 Therefore, a major bottleneck to systematic investigation of normPPIs vs oncoPPIs is having access to appropriate quantities of diverse input materials from patients. Besides the technical challenges, the lack of coordinated efforts between cancer biologists and other disciplines such as proteomics has been a hurdle. Together, these obstacles have slowed systematic investigations of normPPI and oncoPPI networks.

4. ESTABLISHING WORKFLOWS FOR IDENTIFICATION OF ONCOGENIC NETWORKS

We propose a three‐phase strategy to systematically characterise oncoPPIs in human cancers (Figure 2).

FIGURE 2.

FIGURE 2

A cancer‐focused human PPI workflow. A flowchart of the recommended procedures for systematic analysis of normPPIs and oncoPPIs. In Phase I, patient‐derived tumour and normal tissue samples are used to generate cancer and normal cell lines as well as organoids; liver and pancreas have been depicted as examples. In Phase II, two different approaches can be chosen. First, a targeted approach where well‐characterised oncoproteins are used to capture interactions in both patient‐derived cancer and normal cells and organoids using AP‐MS and PL‐MS approaches. Second, an alternative approach can be undertaken on a proteome‐wide scale using XL‐MS, which can provide a picture of vicinal proteins within 30 Å. Phase II will define normPPIs and oncoPPIs in a stepwise manner. The novel normPPIs identified herein are reported to interactome repositories but oncoPPIs are further processed to characterise physical interactions. Physically interacting oncoproteins are subjected to structural analysis for rational drug design. In Phase III, a range of drug discovery approaches are employed to find small molecules that inhibit oncogenic interactions. Functional characterisation of drugs can be done in a personalised manner on the organoids from the same patient

4.1. Phase I: Establishment of patient‐derived normal and cancer organoids

Over the past decades, numerous methods have been used to transform a range of patient cancer samples into immortalised cell lines for studies. 84 Tumour‐derived cell lines are regularly used in pharmacogenomics and cancer biology to investigate biological pathways underlying tumour initiation and development. 85 , 86 Integrated analysis of proteogenomics data generated from hundreds of model cell lines have greatly enhanced our understanding of the molecular mechanisms underlying cancer development. 85 Data generated from cell lines complement the large scale cancer genomics data generated from patient samples. 87 Cell lines offer several advantages including ease of maintenance and proliferation in cell culture with low cost and are amenable to high‐throughput screening (HTS) of anticancer therapeutics. Therefore, establishing cell lines from patient samples would provide an important first step in mapping and identifying potential oncoPPI networks (Figure 2).

In addition, patient‐derived organoids, which are three‐dimensional structures with the capacity to self‐organise into “miniorgans” resembling the tissue of origin, have been established in various cancers 88 (Table 1). Importantly, organoids maintain key cellular hierarchy and function, 111 and can genetically and phenotypically recapitulate the host cancer, with which therapeutic responses can potentially be predicted. 112 Their use as a cancer model for studying protein interactions as well as for screening oncoPPI inhibitors has so far been underutilised but show great potential for precision/personalised medicine. Small‐scale studies using patient‐derived colorectal cancer organoids has identified different proteomic signatures in all patients studied, highlighting the importance of PPI characterisation for personalised therapy. 113 In another study, autophagy impairment and disrupted pathways in Paneth cells was studied using mouse tumour organoids using computational PPI networks. 114 Mouse tumour organoids have also been used for functional validation of PPI inhibitors, Poloppin and Poloppin‐II, which inhibit Polo‐like kinase interactions. 115 Although patient‐derived organoids are technically challenging and costly, the ability to gain a more clinically relevant understanding of a patient's tumour outweighs these challenges in the search for effective targeted therapies.

TABLE 1.

Human organoid biobanks with the number of tumour and normal organoids

Tumour site Tumour Normal Reference
Colorectal 22 19 89
32 18 90
52 41 91
Rectum 65 51 92
Pancreas 8 (2) 93
114

(11)

(19 a )

94
39 (10 a ) 95
52 (5) 96
Stomach 37 7 97
20 4 98
46 17 99
7 4 100
10 8 101
43 34 102
Prostate 7 2 a 103
Breast 56 18 104
95 5 105
Oesophagus 15 12 106
10 1 107
Oral mucosa 25 9 108
Endometrium 14

66

2 a

109
Kidney 54 47 110
a

Normal‐like, noncancer organoids that have a normal phenotype but grown from a cancer sample; brackets indicate that organoids were established but could not be propagated long‐term.

4.2. Phase II: Choosing an appropriate mass spectrometry approach for oncoPPI discovery

After establishment of patient‐derived organoids, two different MS‐based proteomics strategies can be utilised to study normPPI and oncoPPI networks: a focused or proteome‐wide approach (Figure 2).

4.2.1. Focused approach

A set of master oncogenes or tumour suppressors are selected for further characterisation in paired patient‐derived normal and cancer cells or organoids (Figure 2). Depending on the protein of interest's function and localisation either AP‐MS or proximity labelling (PL)‐MS can be chosen.

4.2.1.1 | AP‐MS is the method of choice to discover high affinity interactions

AP‐MS is widely used to purify and characterise interactors of a protein of interest from a complex cell lysate mixture. 45 , 81 , 83 In AP‐MS, either an immunoprecipitation‐grade antibody is immobilised onto a solid‐phase support and then is mixed with cell lysates to capture the target protein and its interactors, or the bait protein is either endogenously or ectopically tagged with epitopes (eg, FLAG, HA and myc) and used to capture interactions. 116 , 117 AP‐MS using an epitope‐tagged approach offers an excellent opportunity to undertake deep and region‐specific interactome studies for most proteins. The most comprehensive human interactome, BioPlex 3.0, has been constructed using this approach. 29 In this project, ~15 000 genes were dual‐tagged with FLAG and HA epitopes and stably overexpressed in HEK293 cells. Affinity‐purified complexes were subjected to MS and network analysis. However, such interactome maps will not distinguish normPPIs from oncoPPIs. To do this, the protein of interest should be compared in both cancer and matched normal cell lines or organoids. For example, RAS gene family members HRAS, KRAS and NRAS carry missense mutations or display dysregulated expression patterns in various cancers and convey aberrant interaction networks. 118 , 119 Therefore, affinity purification and interactome studies of RAS family members or their mutants from both cancer and paired normal cells in pancreatic cancer or other cancers can extract more informative interaction networks and oncogenic pathways (Figure 2). Likewise, a different interaction profile might be observed for a panel of well‐known oncoproteins and tumour suppressors. Together, this approach will not only facilitate the comprehensive definition of the oncogenic map of cancer driver proteins in a tissue‐specific manner but will also enable identification of novel tissue‐specific normPPIs that have not been reported before. The novel PPIs can be reported in interaction repositories such as BioGRID.

MS‐based high‐throughput techniques cannot confidently identify direct interactions in a complex cellular milieu because there is always the possibility that interactions may be bridged by auxiliary proteins or by RNA and DNA molecules. 120 Multisubunit protein complexes are typified by the protein of interest directly interacting with one or two proteins within the same complex and indirectly with other subunits of the complex. 121 Therefore, highly enriched proteins need to be further validated for direct interaction using pairwise comparison experiments such as coimmunoprecipitation or biophysical methods. Such biophysical methods include nuclear magnetic resonance (NMR), X‐Ray crystallography, size exclusion chromatography—multiangle laser light scattering (SEC‐MALLS), dynamic light scattering (DLS) and Förster resonance energy transfer (FRET). Direct interaction interfaces which can be validated can then be used for rational drug design (Figure 2).

4.2.1.2 |PL‐MS is the method of choice to discover transient and organelle‐specific interactions

Engineered peroxidase and biotin ligase enzymes are now routinely applied to proximity labelling of protein interactions. 122 , 123 These include proximity‐dependent biotin‐identification (BioID) 124 and ascorbate peroxidase (APEX)‐based methods 125 or derivatives of these. These approaches were initially developed to overcome the limitations of AP‐MS approaches, in which mild lysis conditions may limit protein solubility and therefore accessibility. 123 , 124 Furthermore, weak and transient interactions, especially interactions involved in cell signalling, may remain undetected. 123 , 126 Therefore PL‐MS is very useful to identify highly dynamic interactions that occur in various biological processes such as the cell cycle, transcription and translation initiation, elongation and termination. BioID, the most widely used approach over the past decade utilises engineered BirA enzymes that are covalently linked to a protein of interest to catalyse the conversion of biotin and ATP into highly reactive biotinoyl‐5′‐AMP molecules which bind lysine residues on neighbouring proteins. Proteins within a 10 nm radius are labelled depending on the type of fused BirA enzyme and duration. 127 Most recently, a BioID‐based approach was used to define the interaction map for 4000 proteins within various intracellular compartments 82 highlighting BioID as a powerful tool for profiling PPIs in different organelles and compartments of live cells.

In mammalian cells, most signal transduction pathways are characterised by weak PPIs that typically occur between globular peptide binding domains (PBDs) of proteins with short, disordered peptide stretches. Over 1800 PBDs have been reported, which indicate their diversity and importance in mediating transient interactions. 128 , 129 Dissecting diversity, low binding affinities and the sensitivity of binding properties to minor sequence variation represent major challenges. 128 In cancer, somatic mutations can commonly occur in PBD‐containing proteins and many of these proteins are drug targets. 128 BioID could be an ideal method to study the transient interactome of oncoproteins containing PBDs in different cancers.

A BioID‐based screen for the “undruggable” SOX2 oncoprotein in patient‐derived squamous cell carcinoma tissue identified SOX2 interaction with EP300 as a therapeutic vulnerability. Indeed, EP300 was shown to be a mediator of SOX2 activity and EP300 bromodomain inhibitors suppressed growth in several lung cancer cell lines. 130 Similarly, interrogation of the oncogenic KRAS G12V interactome using BioID coupled to a CRISPR‐Cas9 loss‐of‐function screen revealed phosphatidylinositol phosphate kinase PIP5K1A as the most negatively enriched target. PIP5K1A depletion reduced KRAS‐dependent proliferation and signalling in several pancreatic cancer cell lines, thus providing a potential drug target for the treatment of KRAS‐mutant cancers. 118 Taken together, growing evidence supports a continual role for proximity labelling technology in the development of precision medicine.

4.2.2. Proteome‐wide approach

This alternative approach can be performed within cells using XL‐MS, 131 whereby the lysine residues of physically‐associating proteins or vicinal proteins (<30 Å) are cross‐linked using homobifunctional chemicals. 132 Cross‐linked proteomes are subjected to proteolytic digestion, size exclusion fractionation, and MS analysis to identify the cross‐linked peptides. In contrast to AP‐MS and BioID, XL‐MS can provide a global snapshot of PPIs within cells in a native state. 133 In addition, it provides orthogonal information to structural maps generated by cryogenic electron microscopy (cryo‐EM), revealing the conformations and dynamic topology of the protein complexes. 134 XL‐MS is the only method that can capture structural and conformational changes of proteins on a global scale in response to stimuli. 134 This approach offers opportunities to study and distinguish the dynamic and protein interaction networks of cancer cells compared to normal counterparts on a proteome‐wide manner.

Tumours acquire a chemoresistance phenotype by acquiring new protein interactions and conformations. 135 , 136 The ability to define interaction networks that drive chemoresistance will provide invaluable insights for developing new therapies and strategies for various drug‐resistant cancers. Chemotherapy remains the standard of care treatment for many cancers. Despite significant advancements in cancer therapy, resistance to chemotherapy frequently occurs 137 and is responsible for poor patient outcomes and cancer relapse. XL‐MS has successfully been used to investigate proteome dynamics and interactions in drug‐sensitive and ‐resistant HeLa cells. 138 This supports using the XL‐MS approach to study protein interaction networks in a proteome scale in normal and paired cancer cell lines (Figure 2).

5. PHASE III: DRUG DISCOVERY

The development of new drug candidates is a very complex process and requires cross‐disciplinary research efforts. Until recently, development of PPI‐specific therapeutics was also a difficult assignment. Unlike typical receptor‐ligand interactions involving a distinct binding pocket, PPI interfaces are usually large, flat and featureless. 139 Recent technological advances in the field of cryo‐EM, 20 , 21 high‐resolution MS 140 and an urgent need for effective drugs to treat life‐threatening diseases such as cancer have massively transformed PPI‐focused drug discovery projects. Accordingly, numerous PPI‐specific drugs have entered clinical trial with some already on the market 17 , 22 (Table 2).

TABLE 2.

List of PPI‐specific drugs and their targets in clinical trials

Inhibitor name Disease area Developer Clinical trial phase ID
BCL‐2/Bax
Venetoclax (ABT199) Chronic lymphocytic leukaemia AbbVie Approved in 2016
MDM2/p53
Idasanutlin (RO5503781) Acute myeloid leukaemia Hoffmann‐La Roche Phase I/II (terminated) NCT03850535
KRT 232 (formerly, AMG 232) Chronic myeloid leukaemia Kartos Therapeutics, Inc Phase I/II (recruiting) NCT04835584
NVP‐CGM097 Solid tumour with p53 wild type status Novartis Pharmaceuticals Phase I (completed) NCT01760525
Milademetan (DS‐3032b) Advanced solid tumour, lymphoma Daiichi Sankyo, Inc Phase I (completed) NCT01877382
SAR405838 Neoplasm malignant Sanofi Phase I (completed) NCT01636479
JNJ‐26854165 Neoplasms Johnson & Johnson Pharmaceutical Research & Development, L.L.C. Phase I (completed) NCT00676910
ALRN‐6924 Advanced solid tumours or lymphomas Aileron Therapeutics Phase I/II (completed) NCT02264613
LFA1/ICAM1
Lifitegrast (SAR 1118) Dry eye Lifelong Vision Foundation Phase IV (completed) NCT03451396
XIAP/Caspase 9
Debio 1143 (AT‐406) Squamous cell carcinoma of the head and neck Debiopharm International SA Phase I/II (active, not recruiting) NCT02022098
LCL‐161 Relapsed or refractory multiple myeloma Novartis Phase II (completed) NCT01955434
Birinapant (TL32711) Advanced or metastatic solid tumours TetraLogic Pharmaceuticals Phase I/II (completed) NCT01188499
ASTX‐660 Advanced solid tumours and lymphomas Astex Pharmaceuticals, Inc Phase I/II (recruiting) NCT02503423
GDC‐0917 Refractory solid tumours or lymphoma Genentech, Inc Phase I (completed) NCT01226277
HGS1029 (AEG40826‐2HCl) Advanced solid tumours Human Genome Sciences, Inc Phase I (completed) NCT00708006
Bromodomain/Histone
Apabetalone (RVX‐000222, RVX‐208) Coronary artery disease Resverlogix Corp Phase III (completed) NCT02586155
Molibresib (GSK525762) NUT midline carcinoma GlaxoSmithKline Phase I (completed) NCT01587703
CPI‐0610 Myelofibrosis Constellation Pharmaceuticals Phase I/II (recruiting) NCT02158858
RO6870810 (formerly, TEN‐010) Acute myeloid leukaemia; myelodysplastic syndromes Hoffmann‐La Roche Phase I (completed) NCT02308761
OTX015 (MK‐8628, Birabresib) Haematologic malignancies Oncoethix GmbH Phase I (completed) NCT01713582
Β‐catenin/CBP
PRI‐724 Liver cirrhosis Komagome Hospital Phase I/II (active, not recruiting) NCT03620474
PD‐1/PD‐L1
CA‐170 Prostatic neoplasms Astellas Pharma, Inc Phase II (completed) NCT01288911

Note: This table is not a comprehensive list of the PPI inhibitors on the market or undergoing clinical trials.

Biological molecules (including antibodies, peptides, aptamers and proteins) and small molecules are the major drugs used in preclinical and clinical studies. 22 The specificity and potency of biological agents such as monoclonal antibodies are superior for targeting PPIs. However, antibody‐based drugs can induce the host immune response and their usefulness is mostly limited to surface PPIs. In addition, antibody manufacturing is very complicated because opportunities for pharmacokinetic modifications are limited. 22 In contrast to antibodies, peptides and small molecules are cheaper and quicker to produce and generally do not induce a host immune response. Despite these features, peptides suffer from poor solubility, short half‐life and poor oral bioavailability. However, peptide‐based drugs are considered as a useful starting point for rational design of mimetics or nonpeptide inhibitors. 141 , 142 Traditionally, small molecules have successfully been used to target small interfaces and offer unique advantages such as oral administration and cell permeability, but they have more potential to exhibit side effects. 143

5.1. Strategies to identify lead compounds to target PPI interfaces

Strategies to identify and validate lead compounds to target PPI interfaces have been reviewed extensively elsewhere. 17 , 22 , 143 Hence, we briefly discuss three major platforms here.

5.1.1. High‐throughput screening

Both industry and academia have successfully used HTS technology for screening libraries of different chemical compounds for drug discovery purposes. 144 Lead compounds identified in HTS screens can be further modified to enhance the specificity and affinity in binding to their target. Compounds identified in HTS screens have been shown to best target PPIs mediated through helix‐groove interactions, where a helical region of a protein binds to the groove of the second protein (eg, p53‐MDM2 complex 145 ). Recent advancements in technology and screening have also identified binders for nonhelix‐groove interactions. For example, the XIAP‐caspase‐9 complex involved in early‐stage regulation of the apoptosis cascade is an extended nonhelical‐groove interaction and targeted by small molecules discovered from HTS studies (Table 2). 146 , 147

5.1.2. In silico screening

Computational methods are increasingly used in drug discovery programs. Here, machine learning and computer‐aided tools are employed to screen chemical libraries for biological targets to find the best hits from millions of compounds. 148 One such approach is pharmacophore‐based virtual scanning where steric and electrochemical factors are mapped in 3D space to provide a simple means to virtually screen compound libraries. Therefore, this approach is used as a foundation to find a hit based on pharmacophoric elements. For PPIs, the pharmacophore represents the molecular features of the protein domains directly involved in the interaction.

5.1.3. Fragment‐based drug discovery

The fragment‐based drug discovery (FBDD) method has successfully been employed for intractable biological targets such as PPI interfaces. 149 , 150 FBDD is a powerful approach to develop effective small molecule compounds starting from weakly binding fragments. In this method, fragment hits (100‐300 Da) with low affinity for the target protein are identified by screening libraries of synthetic or natural product low molecular weight (<300 Da) compounds. 18 , 151 , 152 Through chemical modifications, the identified fragment hits are further developed into drug‐like molecules with higher affinity. 149 , 151 , 152 Recently, dozens of FBDD‐based drugs have entered clinical trials and two have received Food and Drug Administration approval. 153 Using FBDD, PPI inhibitors for oncogenic interfaces such as XIAP‐caspase 9, 154 and RAD51‐BRCA2 155 complexes have been developed (Table 2).

6. CONCLUDING REMARKS

Cancer patients exhibit widespread tumour heterogeneity, which drives an urgent need for precision and personalised treatments. Genomic characterisation of human tumours has led to the generation of vast amounts of high‐resolution sequencing data. These data have enabled identification of many cancer‐driver genes that are involved in the initiation and progression of tumour cells. However, it remains a challenging task to determine how these genomic signatures can be exploited to develop new therapies. Most cancer‐driver genes or oncogenes, discovered to date, are essential for cell growth and maintenance pathways in both healthy and disease states. Therefore, exploiting them as new drug targets may result in adverse effects by perturbing normal physiological pathways. Given that proteins exert their function mostly through interaction with other proteins, it is highly plausible that targeting cancer‐specific oncoPPIs could be a more advantageous approach which may result in a better clinical outcome with minimal side effects. Thus, a focus on generating a comprehensive map of the oncoPPI networks for various human cancers will stimulate targeted drug discovery efforts and will shed more insight into protein networks governing tumour growth.

CONFLICT OF INTEREST

The authors declare no potential conflict of interests.

ACKNOWLEDGEMENTS

Financial support was provided by: National Health & Medical Research Council funding (Investigator Grant #1177305 & Project Grant #1128748 to John E. J. Rasko); Cancer Council NSW project grants (RG14‐05, RG14‐09, RG20‐07, RG20‐12) to John E. J. Rasko and Charles G. Bailey; The Li Ka Shing Foundation to John E. J. Rasko; Tour de Cure research grants to Charles G. Bailey and John E. J. Rasko, and PhD scholarship support to Habib Francis; support grants from Cure The Future Foundation and an anonymous foundation. Open access publishing facilitated by The University of Sydney, as part of the Wiley ‐ The University of Sydney agreement via the Council of Australian University Librarians.

Sharifi Tabar M, Francis H, Yeo D, Bailey CG, Rasko JEJ. Mapping oncogenic protein interactions for precision medicine. Int. J. Cancer. 2022;151(1):7‐19. doi: 10.1002/ijc.33954

Funding information Cancer Council NSW, Grant/Award Numbers: RG14‐05, RG14‐09, RG20‐07, RG20‐12; Cure the Future Foundation; Li Ka Shing Foundation; National Health and Medical Research Council, Grant/Award Numbers: 1128748, 1177305; Tour de Cure

Contributor Information

Charles G. Bailey, Email: c.bailey@centenary.org.au.

John E. J. Rasko, Email: j.rasko@centenary.org.au, @RaskoLab.

REFERENCES

  • 1. Shylasree TS, Bryant A, Athavale R. Chemotherapy and/or radiotherapy in combination with surgery for ovarian carcinosarcoma. Cochrane Database Syst Rev. 2013;(2):CD006246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Butts C, Socinski MA, Mitchell PL, et al. Tecemotide (L‐BLP25) versus placebo after chemoradiotherapy for stage III non‐small‐cell lung cancer (START): a randomised, double‐blind, phase 3 trial. Lancet Oncol. 2014;15(1):59‐68. [DOI] [PubMed] [Google Scholar]
  • 3. Chovanec M, Lauritsen J, Bandak M, et al. Late adverse effects and quality of life in survivors of testicular germ cell tumour. Nat Rev Urol. 2021;18(4):227‐245. [DOI] [PubMed] [Google Scholar]
  • 4. Hwang SY, Chang SJ, Park BW. Does chemotherapy really affect the quality of life of women with breast cancer? J Breast Cancer. 2013;16(2):229‐235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Robertson MJ, Meyerowitz JG, Skiniotis G. Drug discovery in the era of cryo‐electron microscopy. Trends Biochem Sci. 2021;47:124‐135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Llovet JM, Montal R, Sia D, Finn RS. Molecular therapies and precision medicine for hepatocellular carcinoma. Nat Rev Clin Oncol. 2018;15(10):599‐616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Zhang G, Smith SG, Zhou MM. Discovery of chemical inhibitors of human bromodomains. Chem Rev. 2015;115(21):11625‐11668. [DOI] [PubMed] [Google Scholar]
  • 8. Chen X, Liu P, Wang Q, et al. DCZ3112, a novel Hsp90 inhibitor, exerts potent antitumor activity against HER2‐positive breast cancer through disruption of Hsp90‐Cdc37 interaction. Cancer Lett. 2018;434:70‐80. [DOI] [PubMed] [Google Scholar]
  • 9. Bum‐Erdene K, Zhou D, Gonzalez‐Gutierrez G, et al. Small‐molecule covalent modification of conserved cysteine leads to allosteric inhibition of the TEADYap protein‐protein interaction. Cell Chem Biol. 2019;26(3):378‐89 e13. [DOI] [PubMed] [Google Scholar]
  • 10. Momand J, Zambetti GP, Olson DC, George D, Levine AJ. The mdm‐2 oncogene product forms a complex with the p53 protein and inhibits p53‐mediated transactivation. Cell. 1992;69(7):1237‐1245. [DOI] [PubMed] [Google Scholar]
  • 11. Li Z, Ivanov AA, Su R, et al. The OncoPPi network of cancer‐focused protein‐protein interactions to inform biological insights and therapeutic strategies. Nat Commun. 2017;8:14356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Cheng SS, Yang GJ, Wang W, Leung CH, Ma DL. The design and development of covalent protein‐protein interaction inhibitors for cancer treatment. J Hematol Oncol. 2020;13(1):26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Gambini L, Baggio C, Udompholkul P, et al. Covalent inhibitors of protein‐protein interactions targeting lysine, tyrosine, or histidine residues. J Med Chem. 2019;62(11):5616‐5627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Ran X, Gestwicki JE. Inhibitors of protein‐protein interactions (PPIs): an analysis of scaffold choices and buried surface area. Curr Opin Chem Biol. 2018;44:75‐86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Ivanov AA, Gonzalez‐Pecchi V, Khuri LF, et al. OncoPPi‐informed discovery of mitogen‐activated protein kinase kinase 3 as a novel binding partner of c‐Myc. Oncogene. 2017;36(42):5852‐5860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Nevola L, Giralt E. Modulating protein‐protein interactions: the potential of peptides. Chem Commun (Camb). 2015;51(16):3302‐3315. [DOI] [PubMed] [Google Scholar]
  • 17. Nero TL, Morton CJ, Holien JK, Wielens J, Parker MW. Oncogenic protein interfaces: small molecules, big challenges. Nat Rev Cancer. 2014;14(4):248‐262. [DOI] [PubMed] [Google Scholar]
  • 18. Ivanov AA, Khuri FR, Fu H. Targeting protein‐protein interactions as an anticancer strategy. Trends Pharmacol Sci. 2013;34(7):393‐400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Li Z, Ivanov AA, Su R, et al. Corrigendum: the OncoPPi network of cancer‐focused protein‐protein interactions to inform biological insights and therapeutic strategies. Nat Commun. 2017;8:15350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Van Drie JH, Tong L. Cryo‐EM as a powerful tool for drug discovery. Bioorg Med Chem Lett. 2020;30(22):127524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Renaud JP, Chari A, Ciferri C, et al. Cryo‐EM in drug discovery: achievements, limitations and prospects. Nat Rev Drug Discov. 2018;17(7):471‐492. [DOI] [PubMed] [Google Scholar]
  • 22. Lu H, Zhou Q, He J, et al. Recent advances in the development of protein‐protein interactions modulators: mechanisms and clinical trials. Signal Transduct Target Ther. 2020;5(1):213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Mullard A. Pioneering apoptosis‐targeted cancer drug poised for FDA approval. Nat Rev Drug Discov. 2016;15(3):147‐149. [DOI] [PubMed] [Google Scholar]
  • 24. Balmain A. Cancer as a complex genetic trait: tumor susceptibility in humans and mouse models. Cell. 2002;108(2):145‐152. [DOI] [PubMed] [Google Scholar]
  • 25. Eckhardt M, Zhang W, Gross AM, et al. Multiple routes to oncogenesis are promoted by the human papillomavirus‐host protein network. Cancer Discov. 2018;8(11):1474‐1489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Vidal M, Cusick ME, Barabasi AL. Interactome networks and human disease. Cell. 2011;144(6):986‐998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Richards AL, Eckhardt M, Krogan NJ. Mass spectrometry‐based protein‐protein interaction networks for the study of human diseases. Mol Syst Biol. 2021;17(1):e8792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Oughtred R, Rust J, Chang C, et al. The BioGRID database: a comprehensive biomedical resource of curated protein, genetic, and chemical interactions. Protein Sci. 2021;30(1):187‐200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Huttlin EL, Bruckner RJ, Navarrete‐Perea J, et al. Dual proteome‐scale networks reveal cell‐specific remodeling of the human interactome. Cell. 2021;184(11):3022‐40 e28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Kerrien S, Aranda B, Breuza L, et al. The IntAct molecular interaction database in 2012. Nucleic Acids Res. 2012;40(Database issue):D841‐D846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Chatr‐aryamontri A, Ceol A, Palazzi LM, et al. MINT: the molecular INTeraction database. Nucleic Acids Res. 2007;35(Database issue):D572‐D574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Alanis‐Lobato G, Andrade‐Navarro MA, Schaefer MH. HIPPIE v2.0: enhancing meaningfulness and reliability of protein‐protein interaction networks. Nucleic Acids Res. 2017;45(D1):D408‐D414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Du Y, Cai M, Xing X, Ji J, Yang E, Wu J. PINA 3.0: mining cancer interactome. Nucleic Acids Res. 2021;49(D1):D1351‐D1357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Davis RJ. Molecular Analysis of the Chromosomal Translocations Associated with the Pediatric Solid Tumor Alveolar Rhabdomyosarcoma. Penn Libraries, University of Pennsylvania; 1997. https://repository.upenn.edu/dissertations/AAI9800855/ [Google Scholar]
  • 35. Bailey MH, Tokheim C, Porta‐Pardo E, et al. Comprehensive characterization of cancer driver genes and mutations. Cell. 2018;173(2):371‐85 e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Consortium ITP‐CAoWG . Pan‐cancer analysis of whole genomes. Nature. 2020;578(7793):82‐93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Rodriguez‐Martin B, Alvarez EG, Baez‐Ortega A, et al. Pan‐cancer analysis of whole genomes identifies driver rearrangements promoted by LINE‐1 retrotransposition. Nat Genet. 2020;52(3):306‐319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Ellis MJ, Gillette M, Carr SA, et al. Connecting genomic alterations to cancer biology with proteomics: the NCI Clinical Proteomic Tumor Analysis Consortium. Cancer Discov. 2013;3(10):1108‐1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. International Cancer Genome C , Hudson TJ, Anderson W, et al. International network of cancer genome projects. Nature. 2010;464(7291):993‐998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Cheng F, Zhao J, Wang Y, et al. Comprehensive characterization of protein‐protein interactions perturbed by disease mutations. Nat Genet. 2021;53(3):342‐353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Sahni N, Yi S, Taipale M, et al. Widespread macromolecular interaction perturbations in human genetic disorders. Cell. 2015;161(3):647‐660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Sahni N, Yi S, Zhong Q, et al. Edgotype: a fundamental link between genotype and phenotype. Curr Opin Genet dev. 2013;23(6):649‐657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Zhong Q, Simonis N, Li QR, et al. Edgetic perturbation models of human inherited disorders. Mol Syst Biol. 2009;5:321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Rodrigues CHM, Myung Y, Pires DEV, Ascher DB. mCSM‐PPI2: predicting the effects of mutations on protein‐protein interactions. Nucleic Acids Res. 2019;47(W1):W338‐W344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Kamburov A, Lawrence MS, Polak P, et al. Comprehensive assessment of cancer missense mutation clustering in protein structures. Proc Natl Acad Sci USA. 2015;112(40):E5486‐E5495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Chen S, Fragoza R, Klei L, et al. An interactome perturbation framework prioritizes damaging missense mutations for developmental disorders. Nat Genet. 2018;50(7):1032‐1040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Wolfel T, Hauer M, Schneider J, et al. A p16INK4a‐insensitive CDK4 mutant targeted by cytolytic T lymphocytes in a human melanoma. Science. 1995;269(5228):1281‐1284. [DOI] [PubMed] [Google Scholar]
  • 48. Rolland T, Tasan M, Charloteaux B, et al. A proteome‐scale map of the human interactome network. Cell. 2014;159(5):1212‐1226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Nissim S, Leshchiner I, Mancias JD, et al. Mutations in RABL3 alter KRAS prenylation and are associated with hereditary pancreatic cancer. Nat Genet. 2019;51(9):1308‐1314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. O'Connor CM, Leonard D, Wiredja D, et al. Inactivation of PP2A by a recurrent mutation drives resistance to MEK inhibitors. Oncogene. 2020;39(3):703‐717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Haesen D, Abbasi Asbagh L, Derua R, et al. Recurrent PPP2R1A mutations in uterine cancer act through a dominant‐negative mechanism to promote malignant cell growth. Cancer Res. 2016;76(19):5719‐5731. [DOI] [PubMed] [Google Scholar]
  • 52. He B, Gao P, Ding YY, et al. Diverse noncoding mutations contribute to deregulation of cis‐regulatory landscape in pediatric cancers. Sci Adv. 2020;6(30):eaba3064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Rheinbay E, Nielsen MM, Abascal F, et al. Analyses of non‐coding somatic drivers in 2,658 cancer whole genomes. Nature. 2020;578(7793):102‐111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Yuan X, Larsson C, Xu D. Mechanisms underlying the activation of TERT transcription and telomerase activity in human cancer: old actors and new players. Oncogene. 2019;38(34):6172‐6183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Grobner SN, Worst BC, Weischenfeldt J, et al. The landscape of genomic alterations across childhood cancers. Nature. 2018;555(7696):321‐327. [DOI] [PubMed] [Google Scholar]
  • 56. Stern JL, Theodorescu D, Vogelstein B, Papadopoulos N, Cech TR. Mutation of the TERT promoter, switch to active chromatin, and monoallelic TERT expression in multiple cancers. Genes dev. 2015;29(21):2219‐2224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Guo YA, Chang MM, Huang W, et al. Mutation hotspots at CTCF binding sites coupled to chromosomal instability in gastrointestinal cancers. Nat Commun. 2018;9(1):1520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Hnisz D, Weintraub AS, Day DS, et al. Activation of proto‐oncogenes by disruption of chromosome neighborhoods. Science. 2016;351(6280):1454‐1458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Katainen R, Dave K, Pitkanen E, et al. CTCF/cohesin‐binding sites are frequently mutated in cancer. Nat Genet. 2015;47(7):818‐821. [DOI] [PubMed] [Google Scholar]
  • 60. Akdemir KC, Le VT, Chandran S, et al. Disruption of chromatin folding domains by somatic genomic rearrangements in human cancer. Nat Genet. 2020;52(3):294‐305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Surdez D, Zaidi S, Grossetete S, et al. STAG2 mutations alter CTCF‐anchored loop extrusion, reduce cis‐regulatory interactions and EWSR1‐FLI1 activity in Ewing sarcoma. Cancer Cell. 2021;39(6):810‐26 e9. [DOI] [PubMed] [Google Scholar]
  • 62. Bailey CG, Gupta S, Metierre C, et al. Structure‐function relationships explain CTCF zinc finger mutation phenotypes in cancer. Cell Mol Life Sci. 2021;78(23):7519‐7536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Groschel S, Sanders MA, Hoogenboezem R, et al. A single oncogenic enhancer rearrangement causes concomitant EVI1 and GATA2 deregulation in leukemia. Cell. 2014;157(2):369‐381. [DOI] [PubMed] [Google Scholar]
  • 64. Bard‐Chapeau EA, Gunaratne J, Kumar P, et al. EVI1 oncoprotein interacts with a large and complex network of proteins and integrates signals through protein phosphorylation. Proc Natl Acad Sci USA. 2013;110(31):E2885‐E2894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Loukinov DI, Pugacheva E, Vatolin S, et al. BORIS, a novel male germ‐line‐specific protein associated with epigenetic reprogramming events, shares the same 11‐zinc‐finger domain with CTCF, the insulator protein involved in reading imprinting marks in the soma. Proc Natl Acad Sci USA. 2002;99(10):6806‐6811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Debruyne DN, Dries R, Sengupta S, et al. BORIS promotes chromatin regulatory interactions in treatment‐resistant cancer cells. Nature. 2019;572(7771):676‐680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Yang Z, He N, Zhou Q. Brd4 recruits P‐TEFb to chromosomes at late mitosis to promote G1 gene expression and cell cycle progression. Mol Cell Biol. 2008;28(3):967‐976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Luo B, Wu XH, Feng YJ, et al. Nuclear Her2 contributes to paclitaxel resistance in breast cancer cells. Anticancer Drugs. 2021;32(7):709‐716. [DOI] [PubMed] [Google Scholar]
  • 69. Chung I, Reichelt M, Shao L, et al. High cell‐surface density of HER2 deforms cell membranes. Nat Commun. 2016;7:12742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Didiasova M, Schaefer L, Wygrecka M. When place matters: shuttling of Enolase‐1 across cellular compartments. Front Cell Dev Biol. 2019;7:61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Cappello P, Tomaino B, Chiarle R, et al. An integrated humoral and cellular response is elicited in pancreatic cancer by alpha‐enolase, a novel pancreatic ductal adenocarcinoma‐associated antigen. Int J Cancer. 2009;125(3):639‐648. [DOI] [PubMed] [Google Scholar]
  • 72. Pancholi V. Multifunctional alpha‐enolase: its role in diseases. Cell Mol Life Sci. 2001;58(7):902‐920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Kufe DW. MUC1‐C oncoprotein as a target in breast cancer: activation of signaling pathways and therapeutic approaches. Oncogene. 2013;32(9):1073‐1081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Ren J, Bharti A, Raina D, Chen W, Ahmad R, Kufe D. MUC1 oncoprotein is targeted to mitochondria by heregulin‐induced activation of c‐Src and the molecular chaperone HSP90. Oncogene. 2006;25(1):20‐31. [DOI] [PubMed] [Google Scholar]
  • 75. Ren J, Agata N, Chen D, et al. Human MUC1 carcinoma‐associated protein confers resistance to genotoxic anticancer agents. Cancer Cell. 2004;5(2):163‐175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Bitler BG, Goverdhan A, Schroeder JA. MUC1 regulates nuclear localization and function of the epidermal growth factor receptor. J Cell Sci. 2010;123(Pt 10):1716‐1723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Adhikari S, Nice EC, Deutsch EW, et al. A high‐stringency blueprint of the human proteome. Nat Commun. 2020;11(1):5301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Dou Y, Kawaler EA, Cui Zhou D, et al. Proteogenomic characterization of endometrial carcinoma. Cell. 2020;180(4):729‐48 e26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Schmidberger JW, Sharifi Tabar M, Torrado M, et al. The MTA1 subunit of the nucleosome remodeling and deacetylase complex can recruit two copies of RBBP4/7. Protein Sci. 2016;25(8):1472‐1482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Liu F, Rijkers DT, Post H, Heck AJ. Proteome‐wide profiling of protein assemblies by cross‐linking mass spectrometry. Nat Methods. 2015;12(12):1179‐1184. [DOI] [PubMed] [Google Scholar]
  • 81. Sharifi Tabar M, Giardina C, Feng Y, et al. Unique protein interaction networks define the chromatin remodelling module of the NuRD complex. FEBS J. 2022;289:199–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Go CD, Knight JDR, Rajasekharan A, et al. A proximity‐dependent biotinylation map of a human cell. Nature. 2021;595(7865):120‐124. [DOI] [PubMed] [Google Scholar]
  • 83. Sharifi Tabar M, Mackay JP, Low JKK. The stoichiometry and interactome of the nucleosome remodeling and deacetylase (NuRD) complex are conserved across multiple cell lines. FEBS J. 2019;286(11):2043‐2061. [DOI] [PubMed] [Google Scholar]
  • 84. Ramboer E, Vanhaecke T, Rogiers V, Vinken M. Immortalized human hepatic cell lines for in vitro testing and research purposes. Methods Mol Biol. 2015;1250:53‐76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Goodspeed A, Heiser LM, Gray JW, Costello JC. Tumor‐derived cell lines as molecular models of cancer pharmacogenomics. Mol Cancer Res. 2016;14(1):3‐13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Sharma SV, Haber DA, Settleman J. Cell line‐based platforms to evaluate the therapeutic efficacy of candidate anticancer agents. Nat Rev Cancer. 2010;10(4):241‐253. [DOI] [PubMed] [Google Scholar]
  • 87. Hoadley KA, Yau C, Hinoue T, et al. Cell‐of‐origin patterns dominate the molecular classification of 10,000 tumors from 33 types of cancer. Cell. 2018;173(2):291‐304 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Lo YH, Karlsson K, Kuo CJ. Applications of organoids for cancer biology and precision medicine. Nat Cancer. 2020;1(8):761‐773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. van de Wetering M, Francies HE, Francis JM, et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell. 2015;161(4):933‐945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Yan HHN, Siu HC, Ho SL, et al. Organoid cultures of early‐onset colorectal cancers reveal distinct and rare genetic profiles. Gut. 2020;69(12):2165‐2179. [DOI] [PubMed] [Google Scholar]
  • 91. Fujii M, Shimokawa M, Date S, et al. A colorectal tumor organoid library demonstrates progressive loss of niche factor requirements during tumorigenesis. Cell Stem Cell. 2016;18(6):827‐838. [DOI] [PubMed] [Google Scholar]
  • 92. Ganesh K, Wu C, O'Rourke KP, et al. A rectal cancer organoid platform to study individual responses to chemoradiation. Nat Med. 2019;25(10):1607‐1614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Boj SF, Hwang CI, Baker LA, et al. Organoid models of human and mouse ductal pancreatic cancer. Cell. 2015;160(1–2):324‐338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Tiriac H, Belleau P, Engle DD, et al. Organoid profiling identifies common responders to chemotherapy in pancreatic cancer. Cancer Discov. 2018;8(9):1112‐1129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Seino T, Kawasaki S, Shimokawa M, et al. Human pancreatic tumor organoids reveal loss of stem cell niche factor dependence during disease progression. Cell Stem Cell. 2018;22(3):454‐67 e6. [DOI] [PubMed] [Google Scholar]
  • 96. Driehuis E, van Hoeck A, Moore K, et al. Pancreatic cancer organoids recapitulate disease and allow personalized drug screening. Proc Natl Acad Sci USA. 2019;116:26580‐26590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Nanki K, Toshimitsu K, Takano A, et al. Divergent routes toward Wnt and R‐spondin niche independency during human gastric carcinogenesis. Cell. 2018;174(4):856‐69 e17. [DOI] [PubMed] [Google Scholar]
  • 98. Seidlitz T, Merker SR, Rothe A, et al. Human gastric cancer modelling using organoids. Gut. 2019;68(2):207‐217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Yan HHN, Siu HC, Law S, et al. A comprehensive human gastric cancer organoid biobank captures tumor subtype heterogeneity and enables therapeutic screening. Cell Stem Cell. 2018;23(6):882‐97 e11. [DOI] [PubMed] [Google Scholar]
  • 100. Steele NG, Chakrabarti J, Wang J, et al. An organoid‐based preclinical model of human gastric cancer. Cell Mol Gastroenterol Hepatol. 2019;7(1):161‐184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Nuciforo S, Fofana I, Matter MS, et al. Organoid models of human liver cancers derived from tumor needle biopsies. Cell Rep. 2018;24(5):1363‐1376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Mullenders J, de Jongh E, Brousali A, et al. Mouse and human urothelial cancer organoids: a tool for bladder cancer research. Proc Natl Acad Sci USA. 2019;116(10):4567‐4574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Gao D, Vela I, Sboner A, et al. Organoid cultures derived from patients with advanced prostate cancer. Cell. 2014;159(1):176‐187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Kopper O, de Witte CJ, Lohmussaar K, et al. An organoid platform for ovarian cancer captures intra‐ and interpatient heterogeneity. Nat Med. 2019;25(5):838‐849. [DOI] [PubMed] [Google Scholar]
  • 105. Sachs N, de Ligt J, Kopper O, et al. A living biobank of breast cancer organoids captures disease heterogeneity. Cell. 2018;172(1–2):373‐86 e10. [DOI] [PubMed] [Google Scholar]
  • 106. Kijima T, Nakagawa H, Shimonosono M, et al. Three‐dimensional organoids reveal therapy resistance of esophageal and oropharyngeal squamous cell carcinoma cells. Cell Mol Gastroenterol Hepatol. 2019;7(1):73‐91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Li X, Francies HE, Secrier M, et al. Organoid cultures recapitulate esophageal adenocarcinoma heterogeneity providing a model for clonality studies and precision therapeutics. Nat Commun. 2018;9(1):2983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Driehuis E, Kolders S, Spelier S, et al. Oral mucosal organoids as a potential platform for personalized cancer therapy. Cancer Discov. 2019;9(7):852‐871. [DOI] [PubMed] [Google Scholar]
  • 109. Boretto M, Maenhoudt N, Luo X, et al. Patient‐derived organoids from endometrial disease capture clinical heterogeneity and are amenable to drug screening. Nat Cell Biol. 2019;21(8):1041‐1051. [DOI] [PubMed] [Google Scholar]
  • 110. Calandrini C, Schutgens F, Oka R, et al. An organoid biobank for childhood kidney cancers that captures disease and tissue heterogeneity. Nat Commun. 2020;11(1):1310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Clevers H. Modeling development and disease with organoids. Cell. 2016;165(7):1586‐1597. [DOI] [PubMed] [Google Scholar]
  • 112. Drost J, Clevers H. Organoids in cancer research. Nat Rev Cancer. 2018;18(7):407‐418. [DOI] [PubMed] [Google Scholar]
  • 113. Cristobal A, van den Toorn HWP, van de Wetering M, Clevers H, Heck AJR, Mohammed S. Personalized proteome profiles of healthy and tumor human colon organoids reveal both individual diversity and basic features of colorectal cancer. Cell Rep. 2017;18(1):263‐274. [DOI] [PubMed] [Google Scholar]
  • 114. Jones EJ, Matthews ZJ, Gul L, et al. Integrative analysis of Paneth cell proteomic and transcriptomic data from intestinal organoids reveals functional processes dependent on autophagy. Dis Model Mech. 2019;12(3):dmm039511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Narvaez AJ, Ber S, Crooks A, et al. Modulating protein‐protein interactions of the mitotic polo‐like kinases to target mutant KRAS. Cell Chem Biol. 2017;24(8):1017‐28 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Torrado M, Low JKK, Silva APG, et al. Refinement of the subunit interaction network within the nucleosome remodelling and deacetylase (NuRD) complex. FEBS J. 2017;284(24):4216‐4232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Dunham WH, Mullin M, Gingras AC. Affinity‐purification coupled to mass spectrometry: basic principles and strategies. Proteomics. 2012;12(10):1576‐1590. [DOI] [PubMed] [Google Scholar]
  • 118. Adhikari H, Counter CM. Interrogating the protein interactomes of RAS isoforms identifies PIP5K1A as a KRAS‐specific vulnerability. Nat Commun. 2018;9(1):3646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Garcia‐Gomez R, Bustelo XR, Crespo P. Protein‐protein interactions: emerging oncotargets in the RAS‐ERK pathway. Trends Cancer. 2018;4(9):616‐633. [DOI] [PubMed] [Google Scholar]
  • 120. Saldana‐Meyer R, Rodriguez‐Hernaez J, Escobar T, et al. RNA interactions are essential for CTCF‐mediated genome organization. Mol Cell. 2019;76(3):412‐22 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Low JKK, Silva APG, Sharifi Tabar M, et al. The nucleosome remodeling and deacetylase complex has an asymmetric, dynamic, and modular architecture. Cell Rep. 2020;33(9):108450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Yang X, Wen Z, Zhang D, et al. Proximity labeling: an emerging tool for probing in planta molecular interactions. Plant Commun. 2021;2(2):100137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Qin W, Cho KF, Cavanagh PE, Ting AY. Deciphering molecular interactions by proximity labeling. Nat Methods. 2021;18(2):133‐143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Roux KJ, Kim DI, Raida M, Burke B. A promiscuous biotin ligase fusion protein identifies proximal and interacting proteins in mammalian cells. J Cell Biol. 2012;196(6):801‐810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Lam SS, Martell JD, Kamer KJ, et al. Directed evolution of APEX2 for electron microscopy and proximity labeling. Nat Methods. 2015;12(1):51‐54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Wright PE, Dyson HJ. Intrinsically disordered proteins in cellular signalling and regulation. Nat Rev Mol Cell Biol. 2015;16(1):18‐29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Branon TC, Bosch JA, Sanchez AD, et al. Efficient proximity labeling in living cells and organisms with TurboID. Nat Biotechnol. 2018;36(9):880‐887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Cunningham JM, Koytiger G, Sorger PK, AlQuraishi M. Biophysical prediction of protein‐peptide interactions and signaling networks using machine learning. Nat Methods. 2020;17(2):175‐183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Mayer BJ. The discovery of modular binding domains: building blocks of cell signalling. Nat Rev Mol Cell Biol. 2015;16(11):691‐698. [DOI] [PubMed] [Google Scholar]
  • 130. Kim BR, Coyaud E, Laurent EMN, et al. Identification of the SOX2 interactome by BioID reveals EP300 as a mediator of SOX2‐dependent squamous differentiation and lung squamous cell carcinoma growth. Mol Cell Proteomics. 2017;16(10):1864‐1888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Leitner A, Faini M, Stengel F, Aebersold R. Crosslinking and mass spectrometry: an integrated technology to understand the structure and function of molecular machines. Trends Biochem Sci. 2016;41(1):20‐32. [DOI] [PubMed] [Google Scholar]
  • 132. Steigenberger B, Albanese P, Heck AJR, Scheltema RA. To cleave or not to cleave in XL‐MS? J Am Soc Mass Spectrom. 2020;31(2):196‐206. [DOI] [PubMed] [Google Scholar]
  • 133. O'Reilly FJ, Rappsilber J. Cross‐linking mass spectrometry: methods and applications in structural, molecular and systems biology. Nat Struct Mol Biol. 2018;25(11):1000‐1008. [DOI] [PubMed] [Google Scholar]
  • 134. Chavez JD, Bruce JE. Chemical cross‐linking with mass spectrometry: a tool for systems structural biology. Curr Opin Chem Biol. 2019;48:8‐18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Ray Chaudhuri A, Callen E, Ding X, et al. Replication fork stability confers chemoresistance in BRCA‐deficient cells. Nature. 2016;535(7612):382‐387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Wang C, Wu H, Evron T, et al. Structural basis for smoothened receptor modulation and chemoresistance to anticancer drugs. Nat Commun. 2014;5:4355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Gatenby RA, Brown JS. Integrating evolutionary dynamics into cancer therapy. Nat Rev Clin Oncol. 2020;17(11):675‐686. [DOI] [PubMed] [Google Scholar]
  • 138. Chavez JD, Schweppe DK, Eng JK, et al. Quantitative interactome analysis reveals a chemoresistant edgotype. Nat Commun. 2015;6:7928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Wells JA, McClendon CL. Reaching for high‐hanging fruit in drug discovery at protein‐protein interfaces. Nature. 2007;450(7172):1001‐1009. [DOI] [PubMed] [Google Scholar]
  • 140. Aebersold R, Mann M. Mass‐spectrometric exploration of proteome structure and function. Nature. 2016;537(7620):347‐355. [DOI] [PubMed] [Google Scholar]
  • 141. Muttenthaler M, King GF, Adams DJ, Alewood PF. Trends in peptide drug discovery. Nat Rev Drug Discov. 2021;20(4):309‐325. [DOI] [PubMed] [Google Scholar]
  • 142. Lee AC, Harris JL, Khanna KK, Hong JH. A comprehensive review on current advances in peptide drug development and design. Int J Mol Sci. 2019;20(10):2383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Arkin MR, Tang Y, Wells JA. Small‐molecule inhibitors of protein‐protein interactions: progressing toward the reality. Chem Biol. 2014;21(9):1102‐1114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Maculins T, Garcia‐Pardo J, Skenderovic A, et al. Discovery of protein‐protein interaction inhibitors by integrating protein engineering and chemical screening platforms. cell. Chem Biol. 2020;27(11):1441‐51 e7. [DOI] [PubMed] [Google Scholar]
  • 145. Vassilev LT, Vu BT, Graves B, et al. In vivo activation of the p53 pathway by small‐molecule antagonists of MDM2. Science. 2004;303(5659):844‐848. [DOI] [PubMed] [Google Scholar]
  • 146. Oost TK, Sun C, Armstrong RC, et al. Discovery of potent antagonists of the antiapoptotic protein XIAP for the treatment of cancer. J Med Chem. 2004;47(18):4417‐4426. [DOI] [PubMed] [Google Scholar]
  • 147. Feldman T, Kabaleeswaran V, Jang SB, et al. A class of allosteric caspase inhibitors identified by high‐throughput screening. Mol Cell. 2012;47(4):585‐595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Yuriev E, Ramsland PA. Latest developments in molecular docking: 2010‐2011 in review. J Mol Recognit. 2013;26(5):215‐239. [DOI] [PubMed] [Google Scholar]
  • 149. Mullard A. Fragment‐based screening sees the light. Nat Rev Drug Discov. 2020;19(11):742‐743. [DOI] [PubMed] [Google Scholar]
  • 150. Van Molle I, Thomann A, Buckley DL, et al. Dissecting fragment‐based lead discovery at the von Hippel‐Lindau protein:hypoxia inducible factor 1alpha protein‐protein interface. Chem Biol. 2012;19(10):1300‐1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Jhoti H, Williams G, Rees DC, Murray CW. The ‘rule of three’ for fragment‐based drug discovery: where are we now? Nat Rev Drug Discov. 2013;12(8):644‐645. [DOI] [PubMed] [Google Scholar]
  • 152. Murray CW, Rees DC. The rise of fragment‐based drug discovery. Nat Chem. 2009;1(3):187‐192. [DOI] [PubMed] [Google Scholar]
  • 153. Erlanson DA, Fesik SW, Hubbard RE, Jahnke W, Jhoti H. Twenty years on: the impact of fragments on drug discovery. Nat Rev Drug Discov. 2016;15(9):605‐619. [DOI] [PubMed] [Google Scholar]
  • 154. Valkov E, Sharpe T, Marsh M, Greive S, Hyvonen M. Targeting protein‐protein interactions and fragment‐based drug discovery. Top Curr Chem. 2012;317:145‐179. [DOI] [PubMed] [Google Scholar]
  • 155. Scott DE, Coyne AG, Venkitaraman A, Blundell TL, Abell C, Hyvonen M. Small‐molecule inhibitors that target protein‐protein interactions in the RAD51 family of recombinases. ChemMedChem. 2015;10(2):296‐303. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from International Journal of Cancer are provided here courtesy of Wiley

RESOURCES