Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Sep 19.
Published in final edited form as: Curr Opin Chem Biol. 2025 Jun 26;87:102605. doi: 10.1016/j.cbpa.2025.102605

Recent Advances in Chemical Proteomics for Protein Profiling and Targeted Degradation

Peng-Kai Liu 1,a, Zicong Wang 2,a, Lingjun Li 1,2,3,4,5,*
PMCID: PMC12445971  NIHMSID: NIHMS2111048  PMID: 40578074

Abstract

Chemical proteomics has emerged as a powerful approach to decipher protein function, interactions, and targeted degradation pathways in complex biological systems. Recent advances in chemical labeling strategies, including activity-based protein profiling (ABPP), proximity labeling (PL), and proteolysis-targeting chimeras (PROTACs), have facilitated a deeper understanding of protein function and interaction networks. First, ABPP employs covalent probes to selectively label active enzymes, uncovering functional proteomics and drug-target interactions. Innovations such as PhosID-ABPP and streamlined cysteine ABPP have improved site-specific quantification and throughput, enabling proteome-wide analysis of enzyme activity and small-molecule interactions. Second, PL enables the characterization of transient protein–protein interactions using enzymatic or chemically triggered approaches. Advances including TurboID and TransitID enhanced the spatiotemporal resolution of PL. Third, PROTACs expand the scope of targeted protein degradation by leveraging the ubiquitin–proteasome system. Collectively, we highlight recent advancements in integrating mass spectrometry (MS) with these methodologies in the field of chemical proteomics.

Keywords: mass spectrometry, chemical proteomics, activity-based protein profiling (ABPP), proximity labeling (PL), proteolysis-targeting chimeras (PROTACs), protein–protein interactions, drug discovery

Introduction

Mass spectrometry (MS) has emerged as an indispensable tool for identifying and quantifying proteins in complex biological systems [1,2]. Chemical approaches enable selective labeling and enrichment of target proteins, improving the detection of low-abundance species and enhancing the specificity of proteomic analyses [35]. Chemical proteomics employs diverse reaction mechanisms for selective protein labeling: (1) Covalent activity-based reactions target specific amino acid residues, such as cysteine and serine, providing insights into enzyme activity and substrate specificity [6,7]; (2) Bioorthogonal reactions, including copper-catalyzed azide-alkyne cycloaddition (CuAAC) and strain-promoted azide-alkyne cycloaddition (SPAAC), allow protein labeling without interfering with native biological processes [8]; (3) Photoactivation reactions utilize light-activated probes, such as aryl azides and diazirines, to capture dynamic protein interactions [9,10]; and (4) Proximity-dependent biotinylation, such as BioID and TurboID, employs biotin ligases to label and identify the molecular neighborhood of a protein of interest [11,12]. These strategies have significantly advanced the study of protein function and dynamics in biological systems. This review discusses recent applications in integrating MS with these methodologies, including activity-based protein profiling (ABPP), proximity labeling (PL), and proteolysis-targeting chimeras (PROTACs) (Figure 1).

Figure 1. Overview of labeling strategies in chemical proteomics.

Figure 1.

(a) Activity-Based Protein Profiling (ABPP). ABPP utilizes activity-based probes containing a reactive group, a linker, and a tag (e.g., biotin or fluorophore) to selectively label active enzymes. Labeled proteins are subsequently enriched and identified, facilitating studies on drug–target interactions and target visualization. (b) Proximity Labeling (PL). PL employs an engineered enzyme, such as BirA* (a mutant biotin ligase), fused to a bait protein or protein of interest (POI). Upon labeling, nearby proteins become biotinylated and are subsequently enriched for identification, enabling high spatial resolution and intracellular mapping of protein–protein interactions. (c) Proteolysis-Targeting Chimeras (PROTACs). PROTACs are heterobifunctional molecules that induce targeted protein degradation. They contain an E3 ligase ligand, a POI ligand, and a linker, facilitating the formation of a ternary complex with the POI and an E3 ligase. This interaction triggers ubiquitination and subsequent proteasomal degradation of the POI, enabling targeted degradation and overcoming drug resistance.

Affinity-Based Protein Profiling

Activity-based protein profiling (ABPP) has evolved from targeting specific enzyme families into a broader platform for mapping drug and electrophilic compound interactions in biological systems [1316]. Unlike conventional proteomics, ABPP measures protein functionality using activity-based probes (ABPs), which consist of a reactive warhead, a linker, and a reporter group. Under near-physiological conditions, ABPP selectively identifies active proteins in specific experimental or pathological conditions (Figure 1a) [17]. Recent advances in ABPP have improved enrichment and quantification workflows, enabling site-specific and multiplexed proteome-wide analyses. Kleinpenning et al. developed the PhosID approach, which utilizes phosphonate-based capture with immobilized metal affinity chromatography (IMAC) to selectively enrich probe-modified peptides while preserving site information [18]. This enrichment strategy overcomes problems with harsh elution, high non-specific background, and incomplete localization common to traditional biotin pull-downs. Later, Bergen et al. applied PhosID-ABPP to lysates and intact cells, achieving reliable residue-level probe identification [19]. Recently, this workflow was further optimized to illuminate on-target and off-target interactions in kinases, revealing intricate covalent engagements [20]. Collectively, these studies show that phosphonate tagging enables superior site localization compared with biotin-based approaches. However, due to its reliance on metal coordination, strongly acidic, non-phosphorylated peptides can over-bind [21], leading to false positives. Therefore, IMAC conditions must be carefully optimized.

As enrichment strategies have undergone further development, quantitative ABPP methods have evolved to support higher throughput, greater multiplexing, and reduced sample requirements [22]. For instance, isotopic tandem orthogonal proteolysis (isoTOP) ABPP enables single-residue cysteine quantification using isotopically encoded, cleavable tags. However, it requires mass shifts greater than 4 Da to prevent isotopic overlap, which limits multiplexing and necessitates custom probe synthesis [2325]. Similarly, stable isotope labeling by amino acids in cell culture (SILAC) ABPP relies on metabolic labeling with heavy amino acids, restricting its application to cultured cells (Figure 2a) [26]. Although these precursor-ion-based approaches have improved throughput, they are generally limited to duplex or triplex quantification. To address these limitations, product-ion-based quantification using tandem mass tags (TMT/TMTpro) enables the simultaneous analysis of up to 35 samples in a single LC–MS/MS run, significantly boosting throughput while reducing the sample requirement (Figure 2b) [27,28]. SLC-ABPP (streamlined cysteine ABPP) further optimized this process by incorporating an iodoacetamide-based probe for labeling and applying post-proteolysis TMT labeling [29]. Recently, a plate-based single-pot solid-phase-enhanced sample preparation (SP3)-TMT workflow coupled with high-field asymmetric ion mobility spectrometry (FAIMS) enabled the quantification of 18,000–24,000 reactive cysteines from just 10–20 μg of proteome (Figure 2c) [30].

Figure 2. Overview of quantitative ABPP strategies.

Figure 2.

(a) Precursor ion-based quantitative ABPP. SILAC-ABPP utilizes metabolic labeling with heavy and light amino acids, and isoTOP-ABPP employs isotopically encoded probes (>4 Da shift) detectable in MS1 spectra. Quantification relies on extracted ion chromatograms (EIC). (b) Product ion-based quantitative ABPP. Isobaric tags (e.g., TMTpro) enable up to 35-plex multiplexing by introducing a mass reporter and balancer that fragment to release reporter ions in MS/MS scans. This approach enables higher throughput and reduces isotopic overlap compared to precursor ion-based methods. (c) SLC-TMT workflow (using 10 samples as an example). Streamlined cysteine ABPP (SLC-ABPP) labels cysteines at the peptide level following enrichment and digestion, then applies TMT labeling to the resulting peptides. This allows high-throughput screening with advanced MS instrumentation and reduces the amount of sample needed per experiment. (d) sCIP-TMT workflow. Silane-based Cleavable Isotopically labeled Proteomics with TMT (sCIP-TMT) embeds the isobaric label before proteolysis at the protein level, allowing samples to be pooled earlier. This reduces sample-to-sample variability, simplifies downstream sample handling, and preserves the high multiplexing capability for efficiently identifying and quantifying reactive cysteines.

Despite these advances, peptide-level tagging can still suffer from variability arising from the enrichment and enzymatic digestion process. To address this shortcoming, protein-level labeling workflows have been developed to allow earlier incorporation of mass tags (Figure 2d). For instance, sCIP (silane-based cleavable isotopically labeled proteomics) employs a dialkoxydiphenylsilane (DADPS) acid-cleavable linker, which seamlessly incorporates stable isotopes into capture reagents that react with target cysteines at the protein stage [31]. By allowing sample pooling immediately after labeling, sCIP improved quantitative precision over the standard peptide-level workflow and simplifies sample preparation. More recently, sCIP-TMT merges the DADPS-based platform with readily available TMT reagents, alleviating the need for extensive custom syntheses while retaining high multiplex capability [32]. Also, advanced MS techniques, including FAIMS and real-time search-MS3, were applied to effectively filter out interfering ions and mitigate ratio compression effects in quantification [29,30]. Taken together, these methods have enabled ABPP to profile enzyme activities, drug–target engagements, and off-target liabilities at the proteome scale with unprecedented throughput and accuracy [14].

Proximity Labeling

Proximity labeling (PL) has emerged as a powerful approach for characterizing protein–protein interactions by covalently tagging proteins near a bait molecule in living systems [3335]. Unlike traditional affinity purification methods that often lose weak transient interactions during wash steps, PL captures these fleeting contacts under near-physiological conditions. Classic PL methods like BioID use a mutant biotin ligase (BirA*) fused to a bait protein, which generates biotinoyl-5’-AMP intermediates that react with lysine residues on nearby protein (Figure 1b) [11]. However, the large size (~35 kDa) of BirA* and its long labeling time (16–18 hours) limit its effectiveness in capturing transient and rapidly fluctuating interactions [12,36]. To address these challenges, engineered variants such as TurboID and miniTurbo offer faster labeling kinetics and reduced steric interference [12,3639]. TurboID enables rapid tagging within minutes, and miniTurbo, due to its smaller size, minimizes disruption of the bait protein’s function. Beyond biotin ligase-based systems, peroxidase-based methods like APEX and HRP enable rapid covalent tagging of nearby proteins by generating short-lived radicals from biotin-phenol or biotin-tyramide in the presence of hydrogen peroxide. APEX, active in reducing environments, is widely used for intracellular labeling, while HRP, less active in the cytosol, is mainly applied to the cell surface or fixed samples. [12,36]

Building on these advances, Lee et al. developed LOV-Turbo by integrating the light-sensitive LOV domain into the proximity labeling enzyme TurboID, enabling rapid and reversible control of its labeling activity using low-power blue light [40]. In the absence of light, the LOV domain clamps TurboID’s active site, suppressing background labeling. Upon exposure to low-intensity blue light, a rapid conformational change reactivates the enzyme, allowing precise pulse-chase labeling simply by toggling the light. This reversible control makes LOV-Turbo particularly suitable for capturing protein interactions that occur over short timescales or in biotin-rich environments, such as neural systems.

Beyond enzyme engineering, innovative PL strategies have expanded to track protein movement and trafficking. Qin et al. developed TransitID, which employs tandem labeling with two orthogonal enzymes TurboID and engineered ascorbate peroxidase (APEX) to map proteome dynamics in living cells (Figure 3a) [41]. This dual-labeling method distinguishes proteins that traffic between compartments, such as from the cytosol to mitochondria or from the nucleolus to stress granules. For instance, TransitID revealed that stress granules play a protective role by preventing aggregation of the transcription factor JUN during oxidative stress. Furthermore, TransitID was applied to map intercellular protein trafficking, exemplified by its application in profiling communication between tumor cells and macrophages.

Figure 3. Novel proximity labeling (PL) strategies for capturing dynamic proteomes in living systems.

Figure 3.

(a) TransitID: dynamic mapping of proteome trafficking. This dual-enzyme method leverages TurboID (for biotin tagging in a “source” location) and APEX2 (for alkyne-phenol labeling in a “destination” location), enabling spatiotemporal mapping of protein movement within and between cells under near-physiological conditions. Adapted from Ref. [41] with permissions. (b) CAT-S: proximity labeling in primary living samples. In this approach, a targeting photocatalyst and caged thio-quinone methide (thioQM) probe undergo blue-light–driven decaging, releasing a reactive thioQM warhead that tags nearby proteins with biotin in non-genetically modified, difficult-to-transfect samples, including primary mouse tissues and human T cells. Adapted from Ref. [42] with permissions. (c) MultiMap: multiscale PL with tunable resolution. A single photocatalyst (Eosin Y) activates three types of photo-probes (phenol, arylazide, and diazirine), each generating a different labeling radius—from high resolution (~100 Å) to low resolution (~1000 Å). This platform reveals local membrane interactomes on non-engineered live cells or across intercellular synapses, providing multiscale insights into protein–protein networks when paired with biochemical validation and AI-assisted structural analysis. Adapted from Ref. [44] with permissions.

Emerging non-genetic chemical approaches now extend proximity labeling to primary samples that are difficult to manipulate genetically. Liu et al. introduced CAT-S, which enables light-triggered activation of a thio-quinone methide (thioQM) warhead for spatially restricted labeling in hard-to-transfect cells, including native mouse tissues and human T cells (Figure 3b) [42]. Similarly, Takato et al. developed PhoxID, which uses singlet oxygen-mediated photooxidation for rapid PL in genetically unmodified mouse brains, offering unprecedented temporal resolution for receptor-proximal protein mapping [43]. Furthermore, the MultiMap platform enhances PL resolution by employing a single photocatalyst (Eosin Y) to activate different photo probes (diazirine, aryl azide, and phenol derivatives) (Figure 3c) [44]. This allows fine-tuning of labeling radii from ~100 Å for local interactomes to >1000 Å for broader interaction networks. The method’s adjustable resolution allows users to interrogate both local receptor microenvironments and broader intercellular contacts. Deployed in situ on living specimens, MultiMap can capture endogenous plasma-membrane interactomes—even across cell–cell contact zones such as immunological synapses—thereby providing a dynamic blueprint of the cell-surface microenvironment. Moreover, coupling MultiMap data with computational tools like AlphaFold-Multimer provides structural insights into newly identified protein–protein interactions, enriching both proteomic and structural analyses of complex networks. Collectively, these chemistries provide spatial control without genetic manipulation, but they require precise light delivery and thorough evaluation of phototoxicity.

In sum, proximity labeling now enables researchers to genetically encode labeling enzymes (e.g., TurboID, LOV-Turbo, TransitID) or introduce photoactivatable warheads (e.g., CAT-S, PhoxID, MultiMap) for real-time spatial proteome profiling. Taken together, these innovations have cemented proximity labeling as a critical technique in modern chemical proteomics, uncovering dynamic cellular behaviors inaccessible to conventional immunoprecipitation assays.

Proteolysis-targeting chimeras

Mass spectrometry-based proteomics plays a critical role in evaluating PROTAC-mediated degradation, assessing degradation efficiency, and identifying off-target effects. In this section, we discuss the concept of PROTACs and highlight recent chemical proteomics strategies. PROTACs are heterobifunctional molecules that harness the cell’s natural degradation machinery to eliminate disease-causing proteins [45,46]. A PROTAC consists of three key components: a ligand that binds to the protein of interest (POI), a ligand that recruits an E3 ubiquitin ligase, and a linker that connects them (Figure 1c). By simultaneously binding to the POI and the E3 ligase, PROTACs facilitate the formation of a ternary complex, inducing proximity-driven ubiquitination of the POI and marking it for proteasomal degradation [4749]. Unlike traditional inhibitors that merely block catalytic sites, PROTACs selectively degrade target proteins through the ubiquitin–proteasome pathway. As a result, PROTACs can target undruggable proteins, including those that lack well-defined active sites for conventional inhibitors [50]. While PROTACs are highly effective at inducing protein degradation and hold significant therapeutic potential, they often suffer from limited water solubility, poor tissue permeability, and suboptimal pharmacokinetics due to their molecular structure [51,52]. Additionally, challenges such as achieving high specificity, ensuring efficient cellular uptake, and minimizing off-target effects impact delivery strategies and metabolic stability. Future advances in modular and stimuli-responsive PROTAC designs, such as conditionally activatable or tumor-microenvironment-specific PROTACs, could further enhance their clinical applicability [51,52]. Several emerging strategies continue to broaden the landscape of targeted protein manipulation. Autophagy-targeting chimeras (AUTACs) and lysosome-targeting chimeras (LYTACs) redirect proteins to lysosomes via autophagy or receptor-mediated endocytosis, enabling the degradation of cytosolic and membrane proteins that are difficult to target [47]. Additionally, post-translational modification regulators, including PhosTACs and DUBTACs, manipulate protein stability by recruiting phosphatases or deubiquitinases, respectively [53,54]. These approaches expand the chemical biology toolkit beyond the conventional ubiquitin–proteasome pathway, opening new avenues for selective degradation or stabilization of challenging protein targets. In recent years, several innovative platforms—such as antibody-PROTACs, aptamer-PROTACs, and nano-PROTACs—have been developed to address the delivery and pharmacokinetic challenges associated with traditional PROTACs.

Antibody-PROTACs (AbTACs) leverage the high specificity of monoclonal antibodies to direct PROTACs to target cells (Figure 4a) [55,56]. Conjugation to antibodies enables selective recognition of cell surface antigens, followed by receptor-mediated endocytosis and intracellular release of the PROTAC molecule into the cytoplasm. Once inside, the PROTAC binds to the POI and an E3 ubiquitin ligase, facilitating targeted protein degradation via ubiquitin–proteasome system. Recently, Wang et al. developed a ROR1-targeting AbTAC by linking a BRD4-degrading PROTAC to a ROR1 antibody, which demonstrated significant antitumor activity both in vitro and in vivo [57]. Moreover, Cotton et al. developed bispecific AbTAC (Figure 4b) that can simultaneously recruit a membrane-bound E3 ligase and a cell surface POI to induce lysosomal degradation, effectively targeting membrane proteins such as PD-L1 and EGFR [58]. Similarly, aptamer-PROTACs utilize single-stranded DNA or RNA aptamers to improve tumor selectivity and solubility (Figure 4c) [59,60]. Aptamers exhibit low immunogenicity, ease of synthesis, rapid tissue penetration, and high stability, binding targets with high affinity through unique three-dimensional structures. Recently, Liu et al. developed a new archetype of PROTAC (PS-ApTCs), a novel PROTAC platform that utilizes phosphorothioate-modified aptamers for tumor-targeted and spatial-selective degradation of nucleocytoplasmic shuttling proteins, effectively disrupting nucleolin trafficking and enhancing antiproliferative effects [61]. Furthermore, combining PS-ApTCs-mediated nucleolin degradation with aptamer–drug conjugate-based chemotherapy achieves a synergistic tumor inhibition effect.

Figure 4. Overview of engineered PROTAC delivery strategies.

Figure 4.

(a) Antibody-PROTACs (AbTACs). Antibody-PROTAC conjugates selectively bind to antigens on target cells, facilitating endocytosis and PROTAC release. The released PROTAC recruits an E3 ligase, leading to POI ubiquitination and degradation via the ubiquitin–proteasome system (b) Bispecific antibody PROTACs. The bispecific antibody recognizes both the POI and a membrane-bound E3 ligase, inducing POI ubiquitination. The complex undergoes endocytosis, resulting in POI degradation through the lysosomal pathway. (c) Aptamer-based PROTACs. Aptamer-PROTAC conjugates use aptamers for POI recognition and E3 ligase recruitment, enabling ubiquitination and subsequent POI degradation via the ubiquitin–proteasome system. (d) Nano-PROTACs. PROTACs are encapsulated in pH- and cathepsin B-responsive nanoparticles (PSRNs). Upon exposure to acidic conditions and cathepsin B in the tumor microenvironment, the PROTACs are released, facilitating POI degradation.

In addition to these targeted approaches, another common design strategy is the incorporation of nanomaterials into delivery systems. Nano-PROTACs have emerged as a promising approach by conjugating PROTACs with nanomaterials to overcome conventional limitations by improving water solubility, tissue permeability, and metabolic stability [62,63]. Conjugating PROTAC molecules with nanomaterials allows for the utilization of nanoscale delivery systems to enhance the therapeutic efficacy of targeted protein degradation [64]. These systems provide precise spatiotemporal control over drug release, enabling the on-demand activation of PROTACs in target tissues while minimizing off-target effects. For example, Yang et al. developed a type of polymer PROTAC conjugated and pH/cathepsin B sequential responsive nanoparticles (PSRNs) for colorectal cancer therapy (Figure 4d) [64]. These nanoparticles utilized a pH-sensitive design and a cathepsin B-cleavable linker to achieve targeted delivery and precise intracellular release of PROTACs, effectively degrading cyclin-dependent kinase 4 and 6 (CDK4/6). This approach enhanced tumor targeting, improved immune checkpoint blockade efficacy, and demonstrated significant anti-tumor outcomes in colorectal cancer models.

Conclusion

Innovative chemical proteomics approaches continue to transform our ability to decode protein function and interactions in complex biological systems. Recent methodological advances, spanning activity-based protein profiling, proximity labeling, and proteolysis-targeting chimeras (PROTACs), have expanded the toolkit for comprehensive protein analysis. High-throughput enrichment strategies, isobaric labeling, and next-generation instrumentation facilitate deeper and proteome-wide investigations. For example, refined protein-level tagging and enrichment techniques enhance enzyme activity profiling, while light-controlled and chemically triggered proximity labeling methods offer improved spatial and temporal resolution for mapping dynamic protein interactions. In parallel, emerging PROTAC-based approaches, including antibody-, aptamer-, and nano-PROTACs, open new avenues for targeted protein degradation with greater therapeutic precision and efficacy. Moreover, the integration of computational tools, such as machine learning-based structural prediction, with advanced analytical techniques like real-time search-MS3, further optimizes data accuracy and throughput.

We believe that the future of chemical proteomics lies in the integrated application of ABPP, PL, and PROTACs, each offering distinct biological insights. ABPP provides direct information on enzyme activity and functional proteoforms, enabling the identification of catalytically active proteins under physiological or pathological conditions. PL enables spatially resolved interactome mapping, capturing transient and low-affinity protein–protein interactions within defined subcellular microenvironments. PROTACs, in turn, enable the selective degradation of target proteins, supporting perturbation-based studies to uncover causal relationships within signaling networks. Together, they constitute a comprehensive toolkit for dissecting proteome dynamics, regulatory mechanisms, and therapeutic vulnerabilities with unprecedented resolution and information. We anticipate that the convergence of these strategies will not only deepen mechanistic insights into proteome dynamics but also accelerate the discovery of novel druggable targets.

Acknowledgements

The preparation of this manuscript was supported, in part, by the National Institutes of Health Grants P41GM108538, R01AG052324, R01AG078794, and R01 DK071801. L.L. would like to acknowledge funding support of NIH funding R21AG065728, and shared instrument grants (NIH-NCRR S10RR029531, S10OD028473, and S10OD025084), as well as funding support from a Vilas Distinguished Achievement Professorship and Charles Melbourne Johnson Professorship with funding provided by the Wisconsin Alumni Research Foundation and University of Wisconsin-Madison School of Pharmacy. Figures and graphical abstract were created with BioRender.com.

Footnotes

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References

  • 1.Aebersold R, Mann M: Mass spectrometry-based proteomics. Nature 2003, 422:198–207. [DOI] [PubMed] [Google Scholar]
  • 2.Aebersold R, Mann M: Mass-spectrometric exploration of proteome structure and function. Nature 2016, 537:347–355. [DOI] [PubMed] [Google Scholar]
  • 3.Wright MH, Sieber SA: Chemical proteomics approaches for identifying the cellular targets of natural products. Nat Prod Rep 2016, 33:681–708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Shiraiwa K, Cheng R, Nonaka H, Tamura T, Hamachi I: Chemical Tools for Endogenous Protein Labeling and Profiling. Cell Chem Biol 2020, 27:970–985. [DOI] [PubMed] [Google Scholar]
  • 5.Zhang N, Wu J, Zheng Q: Chemical proteomics approaches for protein post-translational modification studies. Biochim Biophys Acta (BBA) - Proteins Proteom 2024, 1872:141017. [DOI] [PubMed] [Google Scholar]
  • 6.Fonović M, Bogyo M: Activity-based probes as a tool for functional proteomic analysis of proteases. Expert Rev Proteom 2008, 5:721–730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Weerapana E, Wang C, Simon GM, Richter F, Khare S, Dillon MBD, Bachovchin DA, Mowen K, Baker D, Cravatt BF: Quantitative reactivity profiling predicts functional cysteines in proteomes. Nature 2010, 468:790–795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Sletten EM, Bertozzi CR: Bioorthogonal Chemistry: Fishing for Selectivity in a Sea of Functionality. Angew Chem Int Ed 2009, 48:6974–6998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Burton NR, Kim P, Backus KM: Photoaffinity labelling strategies for mapping the small molecule–protein interactome. Org Biomol Chem 2021, 19:7792–7809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Dubinsky L, Krom BP, Meijler MM: Diazirine based photoaffinity labeling. Bioorg Med Chem 2012, 20:554–570. [DOI] [PubMed] [Google Scholar]
  • 11.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, 196:801–810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Branon TC, Bosch JA, Sanchez AD, Udeshi ND, Svinkina T, Carr SA, Feldman JL, Perrimon N, Ting AY: Efficient proximity labeling in living cells and organisms with TurboID. Nat Biotechnol 2018, 36:880–887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Niphakis MJ, Cravatt BF: Ligand discovery by activity-based protein profiling. Cell Chem Biol 2024, 31:1636–1651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wang S, Tian Y, Wang M, Wang M, Sun G, Sun X: Advanced Activity-Based Protein Profiling Application Strategies for Drug Development. Front Pharmacol 2018, 9:353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Xiao W, Chen Y, Wang C: Quantitative Chemoproteomic Methods for Reactive Cysteinome Profiling. Isr J Chem 2023, 63. [Google Scholar]
  • 16.Wang C, Weerapana E, Blewett MM, Cravatt BF: A chemoproteomic platform to quantitatively map targets of lipid-derived electrophiles. Nat Methods 2014, 11:79–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Weerapana E, Speers AE, Cravatt BF: Tandem orthogonal proteolysis-activity-based protein profiling (TOP-ABPP)—a general method for mapping sites of probe modification in proteomes. Nat Protoc 2007, 2:1414–1425. [DOI] [PubMed] [Google Scholar]
  • 18.Kleinpenning F, Steigenberger B, Wu W, Heck AJR: Fishing for newly synthesized proteins with phosphonate-handles. Nat Commun 2020, 11:3244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.van Bergen W, Hevler JF, Wu W, Baggelaar MP, Heck AJR: Site-Specific Activity-Based Protein Profiling Using Phosphonate Handles. Mol Cell Proteom 2023, 22:100455. [DOI] [PMC free article] [PubMed] [Google Scholar]; * This study introduces PhosID–ABPP, which uses phosphonate affinity tags to enhance activity-based protein profiling. It enables precise, site-specific mapping of drug binding sites via efficient peptide enrichment under mild conditions, overcoming the limitations of biotin-based methods.
  • 20.Bergen W van Nederstigt AE, Heck AJR, Baggelaar MP: Site-Specific Competitive Kinase Inhibitor Target Profiling Using Phosphonate Affinity Tags. Mol Cell Proteom 2025, doi: 10.1016/j.mcpro.2025.100906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Thingholm TE, Jensen ON, Robinson PJ, Larsen MR: SIMAC (Sequential Elution from IMAC), a Phosphoproteomics Strategy for the Rapid Separation of Monophosphorylated from Multiply Phosphorylated Peptides*. Mol Cell Proteom 2008, 7:661–671. [DOI] [PubMed] [Google Scholar]
  • 22.Chen X, Wong YK, Wang J, Zhang J, Lee Y, Shen H, Lin Q, Hua Z: Target identification with quantitative activity based protein profiling (ABPP). PROTEOMICS 2017, 17. [DOI] [PubMed] [Google Scholar]
  • 23.Hacker SM, Backus KM, Lazear MR, Forli S, Correia BE, Cravatt BF: Global profiling of lysine reactivity and ligandability in the human proteome. Nat Chem 2017, 9:1181–1190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Wang Z, Liu P-K, Li L: A Tutorial Review of Labeling Methods in Mass Spectrometry-Based Quantitative Proteomics. ACS Meas Sci Au 2024, 4:315–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wang Z, Zhang J, Li L: Recent Advances in Labeling-Based Quantitative Glycomics: From High-Throughput Quantification to Structural Elucidation. PROTEOMICS 2025, 25:e202400057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Lanning BR, Whitby LR, Dix MM, Douhan J, Gilbert AM, Hett EC, Johnson TO, Joslyn C, Kath JC, Niessen S, et al. : A road map to evaluate the proteome-wide selectivity of covalent kinase inhibitors. Nat Chem Biol 2014, 10:760–767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Li J, Vranken JGV, Vaites LP, Schweppe DK, Huttlin EL, Etienne C, Nandhikonda P, Viner R, Robitaille AM, Thompson AH, et al. : TMTpro reagents: a set of isobaric labeling mass tags enables simultaneous proteome-wide measurements across 16 samples. Nat Methods 2020, 17:399–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Zuniga NR, Frost DC, Kuhn K, Shin M, Whitehouse RL, Wei T-Y, He Y, Dawson SL, Pike I, Bomgarden RD, et al. : Achieving a 35-Plex Tandem Mass Tag Reagent Set through Deuterium Incorporation. J Proteome Res 2024, 23:5153–5165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kuljanin M, Mitchell DC, Schweppe DK, Gikandi AS, Nusinow DP, Bulloch NJ, Vinogradova EV, Wilson DL, Kool ET, Mancias JD, et al. : Reimagining high-throughput profiling of reactive cysteines for cell-based screening of large electrophile libraries. Nat Biotechnol 2021, 39:630–641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Yang K, Whitehouse RL, Dawson SL, Zhang L, Martin JG, Johnson DS, Paulo JA, Gygi SP, Yu Q: Accelerating multiplexed profiling of protein-ligand interactions: High-throughput plate-based reactive cysteine profiling with minimal input. Cell Chem Biol 2024, 31:565–576.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]; * This study introduces an 18plex TMT ABPP platform that profiles reactive cysteines in 96 well plates using just 10–20 μg of proteome. The method quantifies up to 24,000 cysteine sites from 8,274 proteins, enabling high throughput mapping of ligandability and identification of novel drug on-/off targets.
  • 31.Burton NR, Polasky DA, Shikwana F, Ofori S, Yan T, Geiszler DJ, Leprevost F da V, Nesvizhskii AI, Backus KM: Solid-Phase Compatible Silane-Based Cleavable Linker Enables Custom Isobaric Quantitative Chemoproteomics. J Am Chem Soc 2023, 145:21303–21318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Burton NR, Backus KM: Functionalizing tandem mass tags for streamlining click-based quantitative chemoproteomics. Commun Chem 2024, 7:80. [DOI] [PMC free article] [PubMed] [Google Scholar]; *This study introduces sCIP-TMT, a silane-based cleavable linker strategy for ABPP that enables early-stage isobaric labeling and sample pooling. This method reduces sample preparation time and variance while maintaining high proteomic coverage and accurate quantification of cysteine-reactive sites, enhancing throughput for large-scale screening applications
  • 33.Qin W, Cho KF, Cavanagh PE, Ting AY: Deciphering molecular interactions by proximity labeling. Nat Methods 2021, 18:133–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Milione RR, Schell B-B, Douglas CJ, Seath CP: Creative approaches using proximity labeling to gain new biological insights. Trends Biochem Sci 2024, 49:224–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kang M-G, Rhee H-W: Molecular Spatiomics by Proximity Labeling. Acc Chem Res 2022, 55:1411–1422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Cho KF, Branon TC, Udeshi ND, Myers SA, Carr SA, Ting AY: Proximity labeling in mammalian cells with TurboID and split-TurboID. Nat Protoc 2020, 15:3971–3999. [DOI] [PubMed] [Google Scholar]
  • 37.Mair A, Xu S-L, Branon TC, Ting AY, Bergmann DC: Proximity labeling of protein complexes and cell-type-specific organellar proteomes in Arabidopsis enabled by TurboID. eLife 2019, 8:e47864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Li H, Frankenfield AM, Houston R, Sekine S, Hao L: Thiol-Cleavable Biotin for Chemical and Enzymatic Biotinylation and Its Application to Mitochondrial TurboID Proteomics. J Am Soc Mass Spectrom 2021, 32:2358–2365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Shrestha HK, Sun H, Wang J, Peng J: Profiling Mouse Brain Single-Cell-Type Proteomes Via Adeno-Associated Virus-Mediated Proximity Labeling and Mass Spectrometry. Springer; US; 2024:115–132. [DOI] [PubMed] [Google Scholar]
  • 40.Lee S-Y, Cheah JS, Zhao B, Xu C, Roh H, Kim CK, Cho KF, Udeshi ND, Carr SA, Ting AY: Engineered allostery in light-regulated LOV-Turbo enables precise spatiotemporal control of proximity labeling in living cells. Nat Methods 2023, 20:908–917. [DOI] [PMC free article] [PubMed] [Google Scholar]; * This study presents LOV-Turbo, a light-regulated TurboID variant incorporating a LOV domain for near-zero activity in the dark and rapid activation under blue light. This approach enhances spatial and temporal precision in protein labeling, enabling pulse-chase proteomics in living cells while minimizing background.
  • 41.Qin W, Cheah JS, Xu C, Messing J, Freibaum BD, Boeynaems S, Taylor JP, Udeshi ND, Carr SA, Ting AY: Dynamic mapping of proteome trafficking within and between living cells by TransitID. Cell 2023, 186:3307–3324.e30. [DOI] [PMC free article] [PubMed] [Google Scholar]; ** This study introduces TransitID, a tandem PL technique that sequentially employs TurboID for source compartment biotinylation and APEX2 for destination alkyne-phenol labeling. This approach enables unbiased identification of proteins translocating between organelles or distinct cell types during a defined chase period.
  • 42.Liu Z, Guo F, Zhu Y, Qin S, Hou Y, Guo H, Lin F, Chen PR, Fan X: Bioorthogonal photocatalytic proximity labeling in primary living samples. Nat Commun 2024, 15:2712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Takato M, Sakamoto S, Nonaka H, Valor FYT, Tamura T, Hamachi I: Photoproximity labeling of endogenous receptors in the live mouse brain in minutes. Nat Chem Biol 2025, 21:109–119. [DOI] [PubMed] [Google Scholar]
  • 44.Lin Z, Schaefer K, Lui I, Yao Z, Fossati A, Swaney DL, Palar A, Sali A, Wells JA: Multiscale photocatalytic proximity labeling reveals cell surface neighbors on and between cells. Science 2024, 385:eadl5763. [DOI] [PMC free article] [PubMed] [Google Scholar]; ** This study introduces MultiMap, a photocatalytic PL method that utilizes Eosin Y to activate photo-probes with tunable labeling radii. By conjugating Eosin Y to targeting antibodies, MultiMap enables precise mapping of protein neighborhoods on and between cell surfaces, revealing novel receptor interactors and intercellular synapse components with high spatial and temporal resolution.
  • 45.Sakamoto KM, Kim KB, Kumagai A, Mercurio F, Crews CM, Deshaies RJ: Protacs: Chimeric molecules that target proteins to the Skp1–Cullin–F box complex for ubiquitination and degradation. Proc Natl Acad Sci 2001, 98:8554–8559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Békés M, Langley DR, Crews CM: PROTAC targeted protein degraders: the past is prologue. Nat Rev Drug Discov 2022, 21:181–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zhao L, Zhao J, Zhong K, Tong A, Jia D: Targeted protein degradation: mechanisms, strategies and application. Signal Transduct Target Ther 2022, 7:113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Guharoy M, Bhowmick P, Sallam M, Tompa P: Tripartite degrons confer diversity and specificity on regulated protein degradation in the ubiquitin-proteasome system. Nat Commun 2016, 7:10239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Davis C, Spaller BL, Matouschek A: Mechanisms of substrate recognition by the 26S proteasome. Curr Opin Struct Biol 2021, 67:161–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Dogheim GM, Amralla MT: Proteolysis Targeting Chimera (PROTAC) as a promising novel therapeutic modality for the treatment of triple-negative breast cancer (TNBC). Drug Dev Res 2023, 84:629–653. [DOI] [PubMed] [Google Scholar]
  • 51.Wang C, Zhang Y, Chen W, Wu Y, Xing D: New-generation advanced PROTACs as potential therapeutic agents in cancer therapy. Mol Cancer 2024, 23:110. [DOI] [PMC free article] [PubMed] [Google Scholar]; ** This review provides a comprehensive overview of PROTAC strategies and highlights significant advancements in the development of next-generation PROTACs for cancer therapy.
  • 52.Sun H, Yang K, Zhang X, Fu Y, Yarbro J, Wu Z, Chen P-C, Chen T, Peng J: Evaluation of a Pooling Chemoproteomics Strategy with an FDA-Approved Drug Library. Biochemistry 2023, 62:624–632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Chen P-H, Hu Z, An E, Okeke I, Zheng S, Luo X, Gong A, Jaime-Figueroa S, Crews CM: Modulation of Phosphoprotein Activity by Phosphorylation Targeting Chimeras (PhosTACs). ACS Chem Biol 2021, 16:2808–2815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Henning NJ, Boike L, Spradlin JN, Ward CC, Liu G, Zhang E, Belcher BP, Brittain SM, Hesse MJ, Dovala D, et al. : Deubiquitinase-targeting chimeras for targeted protein stabilization. Nat Chem Biol 2022, 18:412–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Maneiro M, Forte N, Shchepinova MM, Kounde CS, Chudasama V, Baker JR, Tate EW: Antibody–PROTAC Conjugates Enable HER2-Dependent Targeted Protein Degradation of BRD4. ACS Chem Biol 2020, 15:1306–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.VanDyke D, Taylor JD, Kaeo KJ, Hunt J, Spangler JB: Biologics-based degraders — an expanding toolkit for targeted-protein degradation. Curr Opin Biotechnol 2022, 78:102807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Wang L, Ke Y, He Q, Paerhati P, Zhuang W, Yue Y, Liu J, Zhang J, Huang L, Yin Q, et al. : A novel ROR1-targeting antibody-PROTAC conjugate promotes BRD4 degradation for solid tumor treatment. Theranostics 2025, 15:1238–1254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Cotton AD, Nguyen DP, Gramespacher JA, Seiple IB, Wells JA: Development of Antibody-Based PROTACs for the Degradation of the Cell-Surface Immune Checkpoint Protein PD-L1. J Am Chem Soc 2021, 143:593–598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Morita Y, Leslie M, Kameyama H, Volk DE, Tanaka T: Aptamer Therapeutics in Cancer: Current and Future. Cancers 2018, 10:80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Zhang Y, Mi D, Chen Y: An instructive attempt on developing aptamer-constructed PROTAC for breast cancer treatment. Mol Ther - Nucleic Acids 2022, 30:351–352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Liu R, Liu Z, Chen M, Xing H, Zhang P, Zhang J: Cooperatively designed aptamer-PROTACs for spatioselective degradation of nucleocytoplasmic shuttling protein for enhanced combinational therapy. Chem Sci 2023, 15:134–145. [DOI] [PMC free article] [PubMed] [Google Scholar]; *This study introduces PS-ApTCs, an aptamer-PROTAC that enables tumor-targeted and spatioselective degradation of nucleocytoplasmic shuttling proteins. PS-ApTCs disrupts nucleocytoplasmic trafficking and enhances antiproliferative and pro-apoptotic effects.
  • 62.Zhong J, Zhao R, Wang Y, Su Y, Lan X: Nano-PROTACs: state of the art and perspectives. Nanoscale 2024, 16:4378–4391. [DOI] [PubMed] [Google Scholar]
  • 63.Song Y, Dong Q-Q, Ni Y-K, Xu X-L, Chen C-X, Chen W: Nano-Proteolysis Targeting Chimeras (Nano-PROTACs) in Cancer Therapy. Int J Nanomed 2024, 19:5739–5761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Yang L, Yang Y, Zhang J, Li M, Yang L, Wang X, Chen M, Zhang H, He B, Wang X, et al. : Sequential responsive nano-PROTACs for precise intracellular delivery and enhanced degradation efficacy in colorectal cancer therapy. Signal Transduct Target Ther 2024, 9:275. [DOI] [PMC free article] [PubMed] [Google Scholar]; * This study develops stimuli-responsive Nano-PROTACs for controlled protein degradation in colorectal cancer. The sequential delivery mechanism enhances efficacy while minimizing systemic toxicity, presenting a promising strategy for targeted cancer therapy.

RESOURCES