Review Article

Targeted protein degradation: therapeutic potential and challenges of PROTACs and molecular glues

Sun Kim1https://orcid.org/0009-0007-9980-2458, Eui-Bae Jeung1,*https://orcid.org/0000-0001-8936-916X
Author Information & Copyright
1Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
*Corresponding author: Eui-Bae Jeung, Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea, Tel: +82-43-261-2397, E-mail: ebjeung@chungbuk.ac.kr

© Research Institute of Veterinary Medicine, Chungbuk National University.

Received: May 23, 2025; Revised: Jun 10, 2025; Accepted: Jun 11, 2025

Abstract

Targeted protein degradation (TPD) is an emerging therapeutic strategy that leverages the natural protein degradation systems of cells to eliminate disease-associated proteins selectively. Unlike traditional small molecule inhibitors, which merely suppress protein activity, TPD degrades target proteins directly, offering a novel approach to addressing undruggable proteins. The two most extensively studied TPD technologies, proteolysis-targeting chimeras (PROTACs) and molecular glues (MGs), utilize the ubiquitin–proteasome system to induce TPD. PROTACs function as bifunctional molecules that recruit an E3 ubiquitin ligase (E3 ligase) to a target protein, leading to its ubiquitination and subsequent degradation, while MGs enhance protein–protein interactions to facilitate ubiquitination and protein clearance. These approaches have shown promising therapeutic potential in treating cancer, neurodegenerative disorders, and autoimmune diseases, with several compounds currently undergoing clinical trials. Despite these advances, challenges such as limited bioavailability, pharmacokinetic constraints, and target selectivity remain obstacles to the widespread application of TPD-based therapies. Recent developments, including the discovery of novel E3 ligases, linker optimization, and AI-driven drug design, have addressed these limitations, paving the way for the next generation of precision-targeted therapeutics. This paper provides a comprehensive overview of the mechanisms, applications, and future directions of PROTACs and MGs in drug discovery, highlighting their potential to revolutionize modern targeted therapy.

Keywords: molecular targeted therapy; proteolysis-targeting chimeras (PROTACs); ubiquitination; proteolysis; ubiquitin-protein ligases

INTRODUCTION

Continuous protein turnover occurs in the human body, wherein various proteins essential for physiological activities, such as hormones and enzymes, are synthesized while unnecessary proteins are degraded. The normal protein metabolism requires a balance between protein synthesis and degradation to maintain cellular homeostasis [1]. Certain diseases, such as cancer and neurodegenerative disorders, arise when specific proteins accumulate excessively or are improperly degraded, leading to pathological consequences [2]. Small molecule inhibitors (SMIs) have been used widely as traditional therapeutic agents to address these issues. SMIs function by inhibiting the activity of target proteins, mitigating disease progression. These inhibitors typically bind to the active site or a specific binding pocket of the protein, suppressing its function [3]. On the other hand, more than 80% of proteins are classified as “undruggable” because of the lack of well-defined binding pockets or active sites, making them challenging targets for SMIs [4]. In addition, a sustained drug concentration must be maintained to ensure therapeutic efficacy because proteins are continuously synthesized. This requirement increases the risk of adverse effects and enhances the likelihood of off-target interactions, potentially leading to unintended biological consequences [5].

Recently, therapeutic strategies have attempted to overcome these limitations using intracellular protein degradation systems to remove abnormal and misfolded proteins. The two major intracellular protein degradation pathways are the ubiquitin–proteasome system (UPS) and the autophagy–lysosome pathway [6]. The UPS functions by tagging target proteins for degradation through ubiquitination. This ubiquitin-tagged protein is then recognized by the proteasome, which then degrades it, ensuring selective protein turnover and maintaining cellular homeostasis [7]. The autophagy-lysosome pathway is a degradation mechanism that breaks down damaged organelles and protein aggregates, including large protein complexes. In this process, an autophagosome forms around the damaged proteins, encapsulating them within a double-membrane vesicle. The autophagosome then fuses with a lysosome, where hydrolytic enzymes degrade the enclosed proteins using ATP, ensuring cellular quality control and homeostasis [8]. The emerging therapeutic approach of targeted protein degradation (TPD), which leverages the intrinsic protein degradation systems of cells, enables the targeting of proteins that are traditionally considered undruggable by conventional SMIs. Unlike transient inhibition, TPD facilitates the complete removal of the target protein, offering sustained efficacy and potential for repeated therapeutic application [9, 10]. In particular, TPD can target proteins located in challenging cellular compartments, such as the extracellular matrix and cell membrane, and traditionally undruggable targets like transcription factors. This expands the scope of future research opportunities and therapeutic development [11]. Currently, various TPD technologies are being actively researched and developed, including proteolysis-targeting chimeras (PROTACs), molecular glues (MGs), autophagy-targeting chimeras (AUTOTACs), lysosome-targeting chimeras (LyTACs), and autophagy-tethering compounds (ATTECs). Among these, PROTACs and MGs have been studied extensively, with several molecules currently undergoing clinical trials [12]. This study focused on PROTACs and MGs, the two most significant approaches in the TPD field that leverage proteasome-mediated degradation. This review comprehensively explains their mechanisms, therapeutic applications, and prospects.

MECHANISM OF TARGETED PROTEIN DEGRADATION

A clear understanding of the ubiquitination process is essential for elucidating the mechanism of TPD based on the UPS. Ubiquitin is a small, evolutionarily conserved 76-amino acid protein found in all eukaryotic cells. Its final amino acid, glycine, forms a covalent bond with the lysine residue of the target proteins, marking them for degradation through the UPS [13]. Ubiquitin undergoes a sequential enzymatic cascade involving three key enzymes to be conjugated to a target protein. First, the ubiquitin-activating enzyme (E1) activates ubiquitin in an ATP-dependent manner. The ubiquitin-conjugating enzyme (E2) then transfers the activated ubiquitin from E1 to either an E3 ubiquitin ligase (E3 ligase) or directly to the substrate protein. Finally, the E3 ligase recognizes and binds to the target protein, facilitating the transfer of ubiquitin from E2 to the lysine residue of the substrate. A polyubiquitin chain is formed through this process, marking the protein for degradation via the proteasome [14]. The ubiquitinated protein is directed to the 26S proteasome through this three-step process, where it undergoes degradation, ensuring proper protein turnover and cellular homeostasis [15]. The E3 ligase is crucial in recognizing and binding to specific substrate proteins, facilitating their ubiquitination. UPS-based TPD strategies leverage these enzymes to selectively degrade target proteins because of the diversity of E3 ligases, enabling precise and efficient protein elimination [6].

First introduced in 2001, PROTACs are heterobifunctional molecules composed of separate ligands for the target protein and E3 ligase, connected by a chemical linker. This design enables the selective ubiquitination and subsequent degradation of the target protein via the UPS [16]. The target-binding ligand binds to the protein, while the E3 ligase ligand recruits an E3 ligase. Once the E3 ligase is recruited, it facilitates the ubiquitination of the target protein, marking it for proteasomal degradation via the UPS [17].

MGs are a class of small molecules that facilitate the interaction between the target protein and an E3 ligase, promoting ubiquitination and subsequent proteasomal degradation of the target protein [18, 19]. The immunomodulatory imide drug (IMiD) class, which is used as a first-line treatment for multiple myeloma, is a representative example of MGs. IMiDs interact directly with the E3 ligase cereblon (CRBN), facilitating the recruitment of neo-substrates, such as the Ikaros (IKZF) family, leading to their ubiquitination and subsequent degradation [20]. The IKZF family consists of transcription factors traditionally considered undruggable by conventional therapies. Nevertheless, IMiDs induce their polyubiquitination and subsequent proteasomal degradation, eliminating these proteins through MGs-mediated mechanisms (Fig. 1) [21].

jbtr-26-2-21-g1
Fig. 1. Mechanistic overview of TPD by PROTACs and MGs. (A) PROTAC-mediated degradation mechanism through CRBN E3 ligase using a heterobifunctional molecule. (B) MG-mediated degradation mechanism using a monovalent small molecule. In both cases, PROTAC- or MG-induced ternary complex formation between the E3 ligase and the POI leads to polyubiquitination by the E2 ligase. The ubiquitin-tagged POI is then recognized and degraded by the proteasome. Graphic created in BioRender (https://BioRender.com/ynak6b6). PROTACs, proteolysis-targeting chimeras; POI, protein of interest; TPD, targeted protein degradation; MGs, molecular glues; CRBN, cereblon.
Download Original Figure

Although MGs stabilize specific protein–protein interactions (PPIs) through a single small molecule, PROTACs are heterobifunctional molecules that induce proteasomal degradation by simultaneously binding a target protein and an E3 ligase [22]. The main differences between PROTACs and MGs are summarized in Table 1. MGs generally have a low molecular weight, high cell membrane permeability, and favorable drug-like properties, making them well-suited for therapeutic applications. In contrast, PROTACs are larger molecules with complex physicochemical properties that can limit their cellular uptake and metabolic stability, posing challenges for drug delivery and bioavailability [23]. The two approaches are complementary, and MGs are often used as ligands for E3 ligases in PROTAC design, enhancing the efficiency of TPD [2426].

Table 1. Key differences between PROTACs and MGs
PROTACs MGs
Mechanism Induces protein degradation via E3 ligase recruitment Enhances PPI
Structure Bifunctional molecule with a linker Small molecule
Discovery Enables rational design Mostly serendipitous
Target Broad (E3 ligase-dependent) and predictable Limited (PPI-dependent) and undetermined
Cell permeability Lower (large size) Higher (small size)
Optimization Challenging (PK issues) Easier (small-molecule properties)

PROTACs, proteolysis-targeting chimeras; MGs, molecular glues; PPI, protein-protein interactions; PK, pharmacokinetic.

Download Excel Table

In recent years, the discovery of small-molecule ligands for E3 ligases has significantly advanced the understanding and application of PROTAC technology. Most small-molecule PROTACs use Von Hippel-Lindau (VHL) and CRBN E3 ligases for TPD [27]. VHL functions as a substrate adaptor within the Cullin 2 E3 ligase complex, playing a crucial role in targeting hypoxia-inducible factor 1-alpha for rapid and efficient degradation [28, 29]. CRBN, a substrate receptor within the Cullin 4A E3 ligase complex, is the second E3 ligase commonly used in the PROTAC design [30, 31]. IMiD thalidomide and its derivatives have been shown to bind to CRBN, facilitating the degradation of various neo-substrate proteins through the UPS [32, 33]. Despite numerous E3 ligases, only 12 (approximately 1.1%) have been incorporated into PROTAC design, highlighting the need for further exploration and expansion of E3 ligase diversity in TPD strategies [34]. Several E3 ligases, including cIAP (cellular inhibitor of apoptosis protein), MDM2 (mouse double minute 2), DCAF11 (DDB1 and CUL4 associated factor 11), DCAF15, DCAF16, FEM1B (protein fem-1 homolog B), and KEAP1 (Kelch-like ECH-associated protein 1), have been reported as potential candidates for PROTAC development, expanding the range of targetable proteins in TPD strategies [3537]. The E3 ligase library has immense untapped potential for future research. Further studies will be needed to explore new E3 ligases for TPD design, along with expanded investigations into diverse target proteins, to enhance the applicability and effectiveness of TPD-based therapies.

THERAPEUTIC APPLICATIONS OF TARGETED PROTEIN DEGRADATION

PROTAC technologies have been actively studied for various protein-related diseases, showing significant therapeutic efficacy in clinical trials. PROTACs offer a novel approach to cancer therapy by selectively degrading proteins essential for cancer cell proliferation and survival. Notable examples include ARV-110 (for prostate cancer), ARV-471 (for breast cancer), and CFT-1946 (targeting BRAF-mutant cancers) [38, 39]. In addition, PROTAC-based strategies are being explored for the removal of pathogenic protein aggregates associated with neurodegenerative diseases, such as Alzheimer’s disease and Parkinson’s disease [35, 40, 41]. Research is also underway to develop PROTACs targeting key proteins involved in immune response regulation, including interleukin-1 receptor-associated kinase 4, Bruton tyrosine kinase (BTK), and IKZF1/3, to modulate the inflammatory processes and provide potential therapeutic options for autoimmune and inflammatory diseases [42, 43]. The E3 ligands used in PROTACs are derived primarily from IMiDs and generally have a molecular weight of less than 300 Da. Among them, (R)-thalidomide, the E3 ligase ligand in ARV-471, has entered Phase 3 clinical trials. Thalidomide derivatives are the most commonly used CRBN ligands in PROTAC design owing to their high efficacy in recruiting E3 ligases for TPD [44]. Currently, PROTAC drugs targeting various proteins, including androgen receptor, estrogen receptor, B-cell lymphoma-extra large, IKZF1/3, and BTK, have entered clinical trial stages, highlighting their potential as novel therapeutic options (Table 2). Despite their promise, PROTAC drug development faces challenges such as complex synthesis, high costs, and regulatory uncertainties. Thus far, no PROTAC-based therapy has received approval from the Food and Drug Administration (FDA) or other regulatory agencies.

Table 2. Current clinical-stage PROTACs
Compound Target protein Indication Reference
ARV-110 AR Metastatic castration-resistant prostate cancer [65]
ARV-766 AR Metastatic castration-resistant prostate cancer [66]
ARV-471 ER Breast cancer (ER+/HER2-) [67]
DT2216 BCL-XL Hematologic malignancies, solid tumors [68]
NX2127 BTK, IKZF1/3 CLL, NHL [69]
NX5948 BTK B-cell malignant tumors and autoimmune diseases [70]
CFT 1946 Mutant BRAF V600E Melanoma, colorectal cancer [71]
CFT 8634 Bromodomain-containing protein 9 Synovial sarcoma [72]
CFT 8919 Mutant EGFR L858R Non-small cell lung cancer [73]
KT-474 IRAK4 Autoimmune diseases [74]

PROTACs, proteolysis-targeting chimeras; AR, androgen receptor; ER, estrogen receptor; BCL-XL, B-cell lymphoma-extra large; BTK, Bruton tyrosine kinase; IKZF, ikaros; CLL, chronic lymphocytic leukemia; NHL, non-hodgkin’s lymphoma; BRAF, B-Raf proto-oncogene, serine/threonine kinase; EGFR, epidermal growth factor receptor; IRAK4, interleukin-1 receptor-associated kinase 4.

Download Excel Table

Because MGs must induce specific PPIs, their discovery is challenging and often unpredictable, making their development more limited than other therapeutic approaches. In addition, MGs cannot be applied universally to all target proteins because they rely on interactions with specific proteins, restricting their broader applicability [23]. As a result, compared to PROTACs, MGs have attracted relatively less research and development attention. Thus far, the only FDA-approved MGs are thalidomide (Thalomid) and its analogs, lenalidomide (Revlimid) and pomalidomide (Pomalyst) [23]. Table 3 provides examples of MGs that have entered clinical trials. Bristol-Myers Squibb (Princeton, NJ, USA) is a leading company in this field, holding a pipeline of MGs through Celgene, which includes CC-92480 (IKZF1/3), CC-90009 (GSPT1), and CC-220 (IKZF1/3) [4547]. The development of PROTAC-based and MG-based novel therapeutics is expanding steadily, and an increasing number of innovative treatments are expected to emerge.

Table 3. Current clinical-stage MGs
Complound Target protein Indication Reference
CC-92480 IKZF1/3 MM [75]
CC-90009 GSPT1 AML, MDS [76]
CC-220 IKZF1/3 Relapsed/refractory MM, NHL, systemic lupus erythematosus. [77]
E7820 RBM39 Relapsed or refractory AML, MDS or chronic myelomonocytic leukemia [78]
CFT 7455 IKZF1/3 MM, NHL [79]
NVP-DKY709 IKZF2 Cancer immunotherapy [80]

MGs, molecular glues; IKZF, ikaros; MM, multiple myeloma; GSPT1, G1 to S phase transition 1; AML, acute myeloid leukemia; MDS, myeloid dysplasia syndrome; NHL, non-Hodgkin’s lymphoma.

Download Excel Table

CHALLENGES AND FUTURE DIRECTIONS

TPD enables the targeting of “undruggable” proteins that were previously inaccessible to conventional therapies. Unlike traditional inhibitors that just suppress protein activity, TPD eliminates the target protein itself, allowing for complete functional removal. This approach can also reduce resistance caused by long-term selective pressure, making it a promising strategy for durable therapeutic effects [48]. In addition, unlike conventional inhibitors, TPD molecules exhibit a catalytic mode of action, enabling a single molecule to degrade multiple target proteins. This catalytic effect allows for therapeutic efficacy at lower concentrations, potentially reducing toxicity and off-target side effects and improving the overall safety profile of TPD-based therapies [6]. Nevertheless, despite their potential, PROTACs and MGs still face certain limitations that must be addressed before they can be adopted widely as therapeutics for various refractory diseases.

PROTACs face several challenges because of their relatively high molecular weight and pharmacokinetic (PK) limitations. These molecules are generally large, exhibit low solubility, and have restricted cell permeability, which can hinder their intracellular delivery. In addition, metabolic instability can lead to a short half-life in vivo, potentially limiting their therapeutic effectiveness [49]. Optimizing the design of the linker and the ligand that binds to the target protein is necessary to overcome these challenges. Enhancing linker properties can improve cell permeability, stability, and PK, while refining ligand selection can enhance the binding affinity and specificity, ultimately increasing the efficacy of PROTAC-based therapeutics [50]. Furthermore, high concentrations of PROTACs can lead to a phenomenon known as the Hook Effect, which paradoxically reduces protein degradation efficiency. This occurs when ternary complex formation between the target protein, E3 ligase, and PROTAC is impaired. Instead of forming a productive ternary complex, excess PROTAC molecules preferentially bind individually to the target protein or E3 ligase, forming an inactive binary complex and reducing degradation efficiency [51]. Careful dose optimization is essential in clinical settings to prevent this. End-point binding free energy calculation can be used to characterize the stabilization and hook effects in PROTAC systems, optimizing the computational conditions to improve the prediction accuracy for rational PROTAC design [52].

Various innovative approaches are being introduced to overcome the limited accessibility of conventional PROTACs. Various innovative PROTAC technologies have been developed to enhance the selectivity for specific tissues and cells, including Ab-PROTAC, which uses antibodies to target cell membrane surface proteins for improved accessibility [53, 54]; pc-PROTAC, a UV-dependent PROTAC activated at specific locations using UV light [55]; CLIPTAC, which addresses cell permeability issues by linking E3 ligases and target proteins through intracellular chemical reactions [56, 57]; and Folate-PROTAC, which selectively delivers PROTACs to cancer cells with high FOLR1 expression [58]. These advances significantly expand the potential applications of PROTAC technology.

The main limitation of MGs is the lack of rational design strategies, making their discovery and development challenging. The effects of MGs are difficult to predict because they must induce PPIs to enable protein degradation or functional regulation. Furthermore, MGs cannot be applied to proteins not engaging in PPIs, limiting their target scope [23]. The rational development of MGs will require new platforms incorporating screening approaches focused on targets and effectors, along with chemical design strategies [59]. A combination of innovative biochemical assays (TR-FRET, AlphaScreen), cell-based screening methods (viability/toxicity analysis, reporter gene assays, NanoBRET), co-immunoprecipitation, proximity labeling techniques (BioID, TurboID, AirID), and computational approaches is crucial to screen and characterize MGs effectively [6062]. New screening technologies and biochemical analysis methods for MG discovery are evolving. AI-based machine learning is expected to play a key role in predicting the MG mechanisms of action and optimizing drug design [63, 64].

CONCLUSION

TPD is an innovative approach that overcomes the limitations of SMIs by directly degrading target proteins, offering a novel therapeutic strategy for disease treatment. Among current TPD technologies, PROTACs and MGs are the most actively researched, showing promise as therapeutic agents for cancer, neurodegenerative diseases, and autoimmune disorders, with some candidates having entered clinical trials. Using E3 ligases to degrade the target proteins, TPD technologies can effectively target undruggable proteins that are difficult to modulate with conventional drugs. Nevertheless, challenges persist, such as PK limitations in PROTACs and target protein restrictions in MGs, necessitating further technological advancements and research efforts to overcome these limitations. The advances in TPD technology are driving the development of new drug discovery platforms that focus on identifying novel E3 ligases, optimizing drug design, and AI-driven drug development. These advances will enable TPD technology to establish itself as a more precise and effective therapeutic approach, playing a crucial role in treating various diseases, including refractory and difficult-to-treat conditions.

Conflict of Interest

No potential conflict of interest relevant to this article was reported.

Acknowledgements

This work was conducted during the research year of Chungbuk National University in 2025.

Ethics Approval

Not applicable.

References

1.

dos Passos PMS, de Correia CRSTB, de Oliveira CAB, Spagnol V, Morales Martins IF, Roberti Teixeira F. Ubiquitin ligases: proteolytic signaling, protein turnover, and disease.In In: Zelanis A, editor.ed Proteolytic signaling in health and disease. Amsterdam: Elsevier. 2022; p p. 11-40

2.

Van Drie JH. Protein folding, protein homeostasis, and cancer. Chin J Cancer. 2011; 30:124-137

3.

Arkin MR, Wells JA. Small-molecule inhibitors of protein–protein interactions: progressing towards the dream. Nat Rev Drug Discov. 2004; 3:301-317

4.

Zhong L, Li Y, Xiong L, Wang W, Wu M, Yuan T, Yang W, Tian C, Miao Z, Wang T, Yang S. Small molecules in targeted cancer therapy: advances, challenges, and future perspectives. Sig Transduct Target Ther. 2021; 6:201

5.

Bedard PL, Hyman DM, Davids MS, Siu LL. Small molecules, big impact: 20 years of targeted therapy in oncology. Lancet. 2020; 395:1078-1088

6.

Zhao L, Zhao J, Zhong K, Tong A, Jia D. Targeted protein degradation: mechanisms, strategies and application. Sig Transduct Target Ther. 2022; 7:113

7.

Nandi D, Tahiliani P, Kumar A, Chandu D. The ubiquitin-proteasome system. J Biosci. 2006; 31:137-155

8.

Korolchuk VI, Menzies FM, Rubinsztein DC. Mechanisms of cross-talk between the ubiquitin-proteasome and autophagy-lysosome systems. FEBS Lett. 2010; 584:1393-1398

9.

Samarasinghe KTG, Crews CM. Targeted protein degradation: a promise for undruggable proteins. Cell Chem Biol. 2021; 28:934-951

10.

Hanzl A, Winter GE. Targeted protein degradation: current and future challenges. Curr Opin Chem Biol. 2020; 56:35-41

11.

Pliatsika D, Blatter C, Riedl R. Targeted protein degradation: current molecular targets, localization, and strategies. Drug Discov Today. 2024; 29:104178

12.

Tsai JM, Nowak RP, Ebert BL, Fischer ES. Targeted protein degradation: from mechanisms to clinic. Nat Rev Mol Cell Biol. 2024; 25:740-757

13.

Hershko A, Ciechanover A. The ubiquitin system. Annu Rev Biochem. 1998; 67:425-479

14.

Pickart CM, Eddins MJ. Ubiquitin: structures, functions, mechanisms. Biochim Biophys Acta Mol Cell Res. 2004; 1695:55-72

15.

Kleiger G, Mayor T. Perilous journey: a tour of the ubiquitin–proteasome system. Trends Cell Biol. 2014; 24:352-359

16.

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

17.

Paiva SL, Crews CM. Targeted protein degradation: elements of PROTAC design. Curr Opin Chem Biol. 2019; 50:111-119

18.

Dewey JA, Delalande C, Azizi SA, Lu V, Antonopoulos D, Babnigg G. Molecular glue discovery: current and future approaches. J Med Chem. 2023; 66:9278-9296

19.

Baek K, Schulman BA. Molecular glue concept solidifies. Nat Chem Biol. 2020; 16:2-3

20.

Li P, Hu X, Fan Z, Sun S, Ran Q, Wei T, Wei P, Jiang Q, Yan J, Yang N, Jia C, Yang T, Mao Y, Cai X, Xu T, Zhao Z, Qian X, Qin W, Zhuang X, Fan F, Xiao J, Zheng Z, Li S. Novel potent molecular glue degraders against broad range of hematological cancer cell lines via multiple neosubstrates degradation. J Hematol Oncol. 2024; 17:77

21.

Cippitelli M, Stabile H, Kosta A, Petillo S, Gismondi A, Santoni A, Fionda C. Role of Aiolos and Ikaros in the antitumor and immunomodulatory activity of IMiDs in multiple myeloma: better to lose than to find them. Int J Mol Sci. 2021; 22:1103

22.

Ramachandran S, Ciulli A. Building ubiquitination machineries: E3 ligase multi-subunit assembly and substrate targeting by PROTACs and molecular glues. Curr Opin Struct Biol. 2021; 67:110-119

23.

Sasso JM, Tenchov R, Wang D, Johnson LS, Wang X, Zhou QA. Molecular glues: the adhesive connecting targeted protein degradation to the clinic. Biochemistry. 2022; 62:601-623

24.

Lu P, Cheng Y, Xue L, Ren X, Xu X, Chen C, Cao L, Li J, Wu Q, Sun S, Hou J, Jia W, Wang W, Ma Y, Jiang Z, Li C, Qi X, Huang N, Han T. Selective degradation of multimeric proteins by TRIM21-based molecular glue and PROTAC degraders. Cell. 2024; 187:7126-7142.E20

25.

Yang Z, Sun Y, Ni Z, Yang C, Tong Y, Liu Y, Li H, Rao Y. Merging PROTAC and molecular glue for degrading BTK and GSPT1 proteins concurrently. Cell Res. 2021; 31:1315-1318

26.

Robinson SA, Co JA, Banik SM. Molecular glues and induced proximity: an evolution of tools and discovery. Cell Chem Biol. 2024; 31:1089-1100

27.

Diehl CJ, Ciulli A. Discovery of small molecule ligands for the von Hippel-Lindau (VHL) E3 ligase and their use as inhibitors and PROTAC degraders. Chem Soc Rev. 2022; 51:8216-8257

28.

Deshaies RJ, Joazeiro CAP. RING domain E3 ubiquitin ligases. Annu Rev Biochem. 2009; 78:399-434

29.

Buckley SM, Aranda-Orgilles B, Strikoudis A, Apostolou E, Loizou E, Moran-Crusio K, Farnsworth VL, Koller AA, Dasgupta R, Silva JC, Stadtfeld M, Hochedlinger K, Chen EI, Aifantis I. Regulation of pluripotency and cellular reprogramming by the ubiquitin-proteasome system. Cell Stem Cell. 2012; 11:783-798

30.

Wang C, Zhang Y, Wu Y, Xing D. Developments of CRBN-based PROTACs as potential therapeutic agents. Eur J Med Chem. 2021; 225:113749

31.

Brodermann MH, Henderson EK, Sellar RS. The emerging role of targeted protein degradation to treat and study cancer. J Pathol. 2024; 263:403-417

32.

Fischer ES, Böhm K, Lydeard JR, Yang H, Stadler MB, Cavadini S, Nagel J, Serluca F, Acker V, Lingaraju GM, Tichkule RB, Schebesta M, Forrester WC, Schirle M, Hassiepen U, Ottl J, Hild M, Beckwith REJ, Harper JW, Jenkins JL, Thomä NH. Structure of the DDB1–CRBN E3 ubiquitin ligase in complex with thalidomide. Nature. 2014; 512:49-53

33.

Yamamoto J, Ito T, Yamaguchi Y, Handa H. Discovery of CRBN as a target of thalidomide: a breakthrough for progress in the development of protein degraders. Chem Soc Rev. 2022; 51:6234-6250

34.

Schapira M, Calabrese MF, Bullock AN, Crews CM. Targeted protein degradation: expanding the toolbox. Nat Rev Drug Discov. 2019; 18:949-963

35.

Békés M, Langley DR, Crews CM. PROTAC targeted protein degraders: the past is prologue. Nat Rev Drug Discov. 2022; 21:181-200

36.

Zhang X, Luukkonen LM, Eissler CL, Crowley VM, Yamashita Y, Schafroth MA, Kikuchi S, Weinstein DS, Symons KT, Nordin BE, Rodriguez JL, Wucherpfennig TG, Bauer LG, Dix MM, Stamos D, Kinsella TM, Simon GM, Baltgalvis KA, Cravatt BF. DCAF11 supports targeted protein degradation by electrophilic proteolysis-targeting chimeras. J Am Chem Soc. 2021; 143:5141-5149

37.

Troup RI, Fallan C, Baud MGJ. Current strategies for the design of PROTAC linkers: a critical review. Explor Target Anti-tumor Ther. 2020; 1:273-312

38.

Hu J, Xu H, Wu T, Zhang C, Shen H, Dong R, Hu Q, Xiang Q, Chai S, Luo G, Chen X, Huang Y, Zhao X, Peng C, Wu X, Lin B, Zhang Y, Xu Y. Discovery of highly potent and efficient CBP/p300 degraders with strong in vivo antitumor activity. J Med Chem. 2024; 67:6952-6986

39.

He X, Weng Z, Zou Y. Progress in the controllability technology of PROTAC. Eur J Med Chem. 2024; 265:116096

40.

Inuzuka H, Liu J, Wei W, Rezaeian AH. PROTAC technology for the treatment of Alzheimer’s disease: advances and perspectives. Acta Mater Med. 2022; 1:24-41

41.

Amirian R, Badrbani MA, Derakhshankhah H, Izadi Z, Shahbazi MA. Targeted protein degradation for the treatment of Parkinson’s disease: advances and future perspective. Biomed Pharmacother. 2023; 166:115408

42.

Zhang C, Sun X, Song P, Rao Y. The application of PROTACs in immune-inflammation diseases. Bioorg Med Chem. 2024; 115:117967

43.

Galla MS, Sharma N, Mishra P, Shankaraiah N. Recent insights of PROTAC developments in inflammation-mediated and autoimmune targets: a critical review. RSC Med Chem. 2024; 15:2585-2600

44.

Yang N, Kong B, Zhu Z, Huang F, Zhang L, Lu T, Chen Y, Zhang Y, Jiang Y. Recent advances in targeted protein degraders as potential therapeutic agents. Mol Divers. 2024; 28:309-333

45.

Hansen JD, Correa M, Nagy MA, Alexander M, Plantevin V, Grant V, Whitefield B, Huang D, Kercher T, Harris R, Narla RK, Leisten J, Tang Y, Moghaddam M, Ebinger K, Piccotti J, Havens CG, Cathers B, Carmichael J, Daniel T, Vessey R, Hamann LG, Leftheris K, Mendy D, Baculi F, LeBrun LA, Khambatta G, Lopez-Girona A. Discovery of CRBN E3 ligase modulator CC-92480 for the treatment of relapsed and refractory multiple myeloma. J Med Chem. 2020; 63:6648-6676

46.

Surka C, Jin L, Mbong N, Lu CC, Jang IS, Rychak E, Mendy D, Clayton T, Tindall E, Hsu C, Fontanillo C, Tran E, Contreras A, Ng SWK, Matyskiela M, Wang K, Chamberlain P, Cathers B, Carmichael J, Hansen J, Wang JCY, Minden MD, Fan J, Pierce DW, Pourdehnad M, Rolfe M, Lopez-Girona A, Dick JE, Lu G. CC-90009, a novel cereblon E3 ligase modulator, targets acute myeloid leukemia blasts and leukemia stem cells. Blood. 2021; 137:661-677

47.

Matyskiela ME, Zhang W, Man HW, Muller G, Khambatta G, Baculi F, Hickman M, LeBrun L, Pagarigan B, Carmel G, Lu CC, Lu G, Riley M, Satoh Y, Schafer P, Daniel TO, Carmichael J, Cathers BE, Chamberlain PP. A cereblon modulator (CC-220) with improved degradation of Ikaros and Aiolos. J Med Chem. 2018; 61:535-542

48.

Xie X, Yu T, Li X, Zhang N, Foster LJ, Peng C, Huang W, He G. Recent advances in targeting the “undruggable” proteins: from drug discovery to clinical trials. Sig Transduct Target Ther. 2023; 8:335

49.

Zeng S, Huang W, Zheng X, Cheng L, Zhang Z, Wang J, Shen Z. Proteolysis targeting chimera (PROTAC) in drug discovery paradigm: recent progress and future challenges. Eur J Med Chem. 2021; 210:112981

50.

Rej RK, Allu SR, Roy J, Acharyya RK, Kiran INC, Addepalli Y, Dhamodharan V. Orally bioavailable proteolysis-targeting chimeras: an innovative approach in the golden era of discovering small-molecule cancer drugs. Pharmaceuticals. 2024; 17:494

51.

Moreau K, Coen M, Zhang AX, Pachl F, Castaldi MP, Dahl G, Boyd H, Scott C, Newham P. Proteolysis-targeting chimeras in drug development: a safety perspective. Br J Pharmacol. 2020; 177:1709-1718

52.

Xu K, Wang Z, Xiang S, Tang R, Deng Q, Ge J, Jiang Z, Yang K, Hou T, Sun H. Characterizing the cooperative effect of PROTAC systems with end-point binding free energy calculation. J Chem Inf Model. 2024; 64:7666-7678

53.

Chen C, Yang Y, Wang Z, Li H, Dong C, Zhang X. Recent advances in pro-PROTAC development to address on-target off-tumor toxicity. J Med Chem. 2023; 66:8428-8440

54.

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

55.

Xue G, Wang K, Zhou D, Zhong H, Pan Z. Light-induced protein degradation with photocaged PROTACs. J Am Chem Soc. 2019; 141:18370-18374

56.

Lebraud H, Wright DJ, Johnson CN, Heightman TD. Protein degradation by in-cell self-assembly of proteolysis targeting chimeras. ACS Cent Sci. 2016; 2:927-934

57.

Wang C, Zheng C, Wang H, Zhang L, Liu Z, Xu P. The state of the art of PROTAC technologies for drug discovery. Eur J Med Chem. 2022; 235:114290

58.

Liu J, Chen H, Liu Y, Shen Y, Meng F, Ümit Kaniskan H, Jin J, Wei W. Cancer selective target degradation by folate-caged PROTACs. J Am Chem Soc. 2021; 143:7380-7387

59.

Frere GA, de Araujo ED, Gunning PT. Emerging mechanisms of targeted protein degradation by molecular glues. Methods Cell Biol. 2022; 169:1-26

60.

Oleinikovas V, Gainza P, Ryckmans T, Fasching B, Thomä NH. From thalidomide to rational molecular glue design for targeted protein degradation. Annu Rev Pharmacol Toxicol. 2024; 64:291-312

61.

Holdgate GA, Bardelle C, Berry SK, Lanne A, Cuomo ME. Screening for molecular glues – challenges and opportunities. SLAS Discov. 2024; 29:100136

62.

Domostegui A, Nieto-Barrado L, Perez-Lopez C, Mayor-Ruiz C. Chasing molecular glue degraders: screening approaches. Chem Soc Rev. 2022; 51:5498-5517

63.

Duran-Frigola M, Cigler M, Winter GE. Advancing targeted protein degradation via multiomics profiling and artificial intelligence. J Am Chem Soc. 2023; 145:2711-2732

64.

Sathe G, Sapkota GP. Proteomic approaches advancing targeted protein degradation. Trends Pharmacol Sci. 2023; 44:786-801

65.

Gao X, Burris HA, Vuky J, Dreicer R, Sartor AO, Sternberg CN, Percent IJ, Hussain MHA, Kalebasty AR, Shen J, Heath EI, Abesada-Terk G, Gandhi SG, McKean M, Lu H, Berghorn E, Gedrich R, Chirnomas SD, Vogelzang NJ, Petrylak DP. Phase 1/2 study of ARV-110, an androgen receptor (AR) PROTAC degrader, in metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol. 2022; 40:17

66.

Petrylak DP, Stewart TF, Gao X, Berghorn E, Lu H, Chan E, Gedrich R, Lang JM, McKean M. A phase 2 expansion study of ARV-766, a PROTAC androgen receptor (AR) degrader, in metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol. 2023; 41:TPS290

67.

Snyder LB, Flanagan JJ, Qian Y, Gough SM, Andreoli M, Bookbinder M, Cadelina G, Bradley J, Rousseau E, Chandler J, Willard R, Pizzano J, Crews CM, Crew AP, Houston J, Moore MD, Peck R, Taylor I. Abstract 44: the discovery of ARV-471, an orally bioavailable estrogen receptor degrading PROTAC for the treatment of patients with breast cancer. Cancer Res. 2021; 81:44

68.

He Y, Koch R, Budamagunta V, Zhang P, Zhang X, Khan S, Thummuri D, Ortiz YT, Zhang X, Lv D, Wiegand JS, Li W, Palmer AC, Zheng G, Weinstock DM, Zhou D. DT2216: a Bcl-xL-specific degrader is highly active against Bcl-xL-dependent T cell lymphomas. J Hematol Oncol. 2020; 13:95

69.

Robbins DW, Kelly A, Tan M, McIntosh J, Wu J, Konst Z, Kato D, Peng G, Mihalic J, Weiss D, Perez L, Tung J, Kolobova A, Borodovsky S, Rountree R, Tenn-McClellan A, Noviski M, Ye J, Basham S, Ingallinera T, McKinnell J, Karr DE, Powers J, Guiducci C, Sands A. Nx-2127, a degrader of BTK and IMiD neosubstrates, for the treatment of B-cell malignancies. Blood. 2020; 136:34

70.

Robbins DW, Noviski M, Rountree R, Tan M, Brathaban N, Ingallinera T, Karr DE, Kelly A, Konst Z, Ma J, Tenn-McClellan A, McKinnell J, Perez L, Guiducci C, Hansen G, Sands A. Nx-5948, a selective degrader of BTK with activity in preclinical models of hematologic and brain malignancies. Blood. 2021; 138:2251

71.

McKean M, Spira AI, Rosen E, Subbiah V, Moreno V, Gambardella V, Vieito M, Saavedra O, Cousin S, Cassier PA, Lobbardi R, Schönborn-Kellenberger O, Hurh E, Ruisi MM, Buchbinder EI. A phase 1/2 study of CFT1946, a novel, bifunctional degradation activating compound (BIDAC) degrader, of mutant BRAF V600 as monotherapy and in combination with trametinib, in mutant BRAF V600 solid tumors. J Clin Oncol. 2023; 41:TPS3163

72.

Poling LL, Cocozziello D, He M, Hurh E, Lobbardi R, Jackson KL, Fisher SL, Pollock RM. CFT8634, a clinical stage BRD9 Bi DAC™ degrader, is active in a subset of multiple myeloma cell line models and synergistic when combined with pomalidomide. Blood. 2023; 142:6594

73.

Park ES, Ahn JY, Baddour J, Chaturvedi P, Isabel Chiu M, Cole KS, Crystal AS, Duplessis M, Fisher SL, Good AC, Hird AW, Huang H, Hulton CH, Ladd B, Michael RE, Nasveschuk CG, Phillips AJ, Pollock RM, Sarkissian G, Rahman MF, Simard JR. Preclinical evaluation of CFT8919 as a mutant selective degrader of EGFR with L858R activating mutations for the treatment of non-small cell lung cancer. Watertown: C4 Therapeutics. 2021

74.

Ackerman L, Acloque G, Bacchelli S, Schwartz H, Feinstein BJ, La Stella P, Alavi A, Gollerkeri A, Davis J, Campbell V, McDonald A, Agarwal S, Karnik R, Shi K, Mishkin A, Culbertson J, Klaus C, Enerson B, Massa V, Kuhn E, Sharma K, Keaney E, Barnes R, Chen D, Zheng X, Rong H, Sabesan V, Ho C, Mainolfi N, Slavin A, Gollob JA. IRAK4 degrader in hidradenitis suppurativa and atopic dermatitis: a phase 1 trial. Nat Med. 2023; 29:3127-3136

75.

Richardson PG, Vangsted AJ, Ramasamy K, Trudel S, Martínez J, Mateos MV, Rodríguez Otero P, Lonial S, Popat R, Oriol A, Karanes C, Orlowski RZ, Berdeja JG, Wong L, Shi C, Lamba M, Barnett E, Pierce DW, Pourdehnad M, Bahlis NJ. First-in-human phase I study of the novel CELMoD agent CC-92480 combined with dexamethasone (DEX) in patients (pts) with relapsed/refractory multiple myeloma (RRMM). J Clin Oncol. 2020; 38:8500

76.

Uy GL, Minden MD, Montesinos P, DeAngelo DJ, Altman JK, Koprivnikar J, Vyas P, Fløisand Y, Vidriales MB, Gjertsen BT, Esteve J, Buchholz TJ, Couto S, Fan J, Hanna B, Li L, Pierce DW, Hege K, Pourdehnad M, Zeidan AM. Clinical activity of CC-90009, a cereblon E3 ligase modulator and first-in-class GSPT1 degrader, as a single agent in patients with relapsed or refractory acute myeloid leukemia (R/R AML): first results from a phase I dose-finding study. Blood. 2019; 134:232

77.

Lonial S, Popat R, Hulin C, Jagannath S, Oriol A, Richardson PG, Facon T, Weisel K, Larsen JT, Minnema MC, Abdallah AO, Badros AZ, Knop S, Stadtmauer EA, Cheng Y, Amatangelo M, Chen M, Nguyen TV, Amin A, Peluso T, van de Donk NWCJ. Iberdomide plus dexamethasone in heavily pretreated late-line relapsed or refractory multiple myeloma (CC-220-MM-001): a multicentre, multicohort, open-label, phase 1/2 trial. Lancet Haematol. 2022; 9:E822-E832

78.

Bewersdorf JP, Stahl M, Taylor J, Mi X, Chandhok NS, Watts J, Derkach A, Wysocki M, Lu SX, Bourcier J, Hogg SJ, Rahman J, Chaudhry S, Totiger TM, Abdel-Wahab O, Stein EM. E7820, an anti-cancer sulfonamide, degrades RBM39 in patients with splicing factor mutant myeloid malignancies: a phase II clinical trial. Leukemia. 2023; 37:2512-2516

79.

Berdeja J, Ailawadhi S, Horwitz SM, Matous JV, Mehta-Shah N, Martin T, Muchtar E, Richardson PG, Richard S, Bhutani M, Yee AJ, Palmer MR, Gorman C, Schoenborn-Kellenberger O, Bilic S, Crystal A, Mahler M, Lonial S. A phase 1 study of CFT7455, a novel degrader of IKZF1/3, in multiple myeloma and non-Hodgkin lymphoma. Blood. 2021; 138:1675

80.

Bonazzi S, d’Hennezel E, Beckwith REJ, Xu L, Fazal A, Magracheva A, Ramesh R, Cernijenko A, Antonakos B, Bhang HC, García Caro R, Cobb JS, Ornelas E, Ma X, Wartchow CA, Clifton MC, Forseth RR, Fortnam BH, Lu H, Csibi A, Tullai J, Carbonneau S, Thomsen NM, Larrow J, Chie-Leon B, Hainzl D, Gu Y, Lu D, Meyer MJ, Alexander D, Kinyamu-Akunda J, Sabatos-Peyton CA, Dales NA, Zécri FJ, Jain RK, Shulok J, Karen Wang Y, Briner K, Porter JA, Tallarico JA, Engelman JA, Dranoff G, Bradner JE, Visser M, Solomon JM. Discovery and characterization of a selective IKZF2 glue degrader for cancer immunotherapy. Cell Chem Biol. 2023; 30:235-247.E12