08/30/2024
TPD/PROTAC as a transformative drug discovery paradigm
Traditional drug development depends upon the discovery of a small molecule that binds to the active site of a protein of interest (POI) and functionally blocks its activity. However, this type of ‘occupancy-driven’ pharmacology is only suitable for a narrow range of applications, as it requires the target to have both a well-defined binding pocket and established enzymatic activity. Consequently, an estimated 80-90% of the human proteome – including transcription factors, scaffolding proteins, and many other clinically relevant targets – is considered ‘undruggable’ by conventional strategies.
Targeted protein degradation (TPD) is a powerful biological modality that overcomes these limitations by leveraging the cell’s endogenous proteolytic machinery to degrade, rather than simply inhibit, a POI. One of the most popular TPD technologies is that of proteolysis-targeting chimeras, or PROTACs. Simply put, a PROTAC consists of two ligands joined by a linker region. While one ligand engages the POI, the other recruits an E3 ubiquitin ligase. Once brought within sufficient proximity, the E3 ligase polyubiquitinates the POI, inducing its degradation by the proteasome. In this way, PROTACs and other TPD strategies physically eliminate target proteins, rather than simply inhibiting their enzymatic function. This is particularly important given that many enzyme families – including kinases and poly(ADP-ribose) polymerases (PARPs) – are known to play critical scaffolding and signaling roles in addition to their catalytic functions (Kim et al., 2023). The advantages of target depletion over inactivation are also highlighted in the context of cancer and treatment resistance. For example, treatment with RAF kinase inhibitors has been shown to promote tumor growth in some contexts by altering RAF localization and interaction patterns even while blocking its enzymatic activity (Hatzivassiliou et al., 2010).
The most transformative aspect of TPD, however, lies in its ‘event-driven’ pharmacology. While conventional ligands must engage with and inhibit a specific active site, binding of a TPD ligand anywhere on the POI is sufficient to induce degradation. As a result, TPD approaches can be readily applied to otherwise-challenging targets, such as proteins with broad, shallow active sites, or those with ‘smooth’ surfaces and few small-molecule binding pockets. Furthermore, because these degraders are catalytic and recyclable (i.e., upon POI degradation, the degrader dissociates and is free to engage with a new target molecule), they can be administered at much lower doses than traditional inhibitor drugs, and less frequent administration is required. TPD approaches are thought to be less susceptible than traditional inhibitors to the development of ‘escaping’ mutations (Lu et al., 2020) and have been successfully used to overcome acquired drug resistance in cancer and viral contexts (Sun et al., 2019; de Wispelaere et al., 2019). At the same time, TPD-based strategies exhibit exquisite specificity and selectivity, as demonstrated by the specific degradation of the transcriptional regulator CDK9 with no effect on other CDK family members (Olson et al., 2018).
Given these advantages, it should come as no surprise that PROTACs and other TPD strategies have attracted significant clinical attention. In 2019, the first PROTAC degraders – ARV-471 and ARV-110 –entered clinical trials, representing a watershed moment for TPD therapeutics. ARV-471, also known as vepdegestrant, is a PROTAC degrader targeting the estrogen receptor (ER), developed by Arvinas and Pfizer to treat patients with ER+/HER2- breast cancer. In a Phase 1/2 study (NCT04072952), vepdegestrant showed promising results, both as a monotherapy and in combination with palbociclib (Hamilton et al., 2022), and Phase 3 trials in patients with advanced metastatic breast cancer are ongoing (NCT05654623; Campone et al., 2023). ARV-110, also known as bavdegalutamide, was developed by Arvinas to target and degrade the androgen receptor (AR) in patients with metastatic castration-resistant prostate cancer (mCRPC) (Gao et al., 2022). Phase 1 trials (NCT03888612) established the drug’s safety and tolerability, while ongoing Phase 2 trials continue to characterize its effectiveness and pharmacodynamics.
Bavdegalutamide has been followed in the clinical pipeline by a number of additional AR-targeting PROTACs, including compounds developed by Arvinas (ARV-766; NCT05067140), BMS/Celgene (CC-94676; NCT04428788), and Hinova (HP518; NCT06155084) (Jia & Han, 2023). Currently in Phase 1 trials (NCT05225584), Kymera Therapeutics has developed a STAT3 degrader, KT-333, for the treatment of relapsed/refractory lymphomas, leukemias, and solid tumors (Starodub et al., 2022). Another promising degrader, NX-2127, was developed by Nurix Therapeutics to target Bruton’s tyrosine kinase (BTK) for the treatment of B-cell malignancies (Mato et al., 2022); following a partial hold, Phase 1 clinical trials were resumed in March 2024 (NCT04830137).
Early PROTAC therapeutics were designed to target well-studied proteins with existing pharmacological inhibitors, enabling their comparative safety and efficacy to be evaluated. Now that their general utility and tolerability has been established, researchers have begun to apply these technologies to novel targets and diseases for which conventional treatments are insufficient or wholly unavailable. In particular, interest has arisen in the application of PROTACs to proteopathies and other neurodegenerative diseases. In February 2024, Arvinas initiated Phase 1 trials of ARV-102, the first PROTAC treatment for neurodegenerative diseases. This drug targets the kinase LRRK2, associated with Parkinson’s disease and progressive supranuclear palsy, and was shown in primate studies to reduce LRRK2 levels in the cortex, striatum, and cerebellum by over 85% (Arvinas, 2024). PROTACs targeting Tau for the treatment of frontotemporal dementia, Alzheimer’s disease, and other tauopathies have also shown positive preclinical results, with the compound QC-01-175 significantly reducing levels of aberrant Tau in patient-derived neurons (Lu et al., 2018; Silva et al., 2022). Other groups have targeted Alpha-synuclein as a treatment for Parkinson’s disease and other synucleinopathies, both alone (Tong et al., 2023) and in combination with Tau (Zhu et al., 2024), mutant HTT protein as a treatment for Huntington’s disease (Li et al., 2019), and TDP-43 as a treatment for amyotrophic lateral sclerosis (ALS) (Tseng et al., 2023).
Another category of innovative degrader-based therapeutics applies PROTAC technology to exogenous targets for the treatment of infectious diseases. The telaprevir-based degrader DGY-08-097, for example, degrades the protease of the hepatitis C virus (HCV) and exhibits potent anti-viral activity, even in cases where mutations have rendered the HCV protease resistant to conventional inhibitors (de Wispelaere et al., 2019). Other groups have leveraged PROTACs against SARS-CoV-2 and other coronaviruses (Desantis et al., 2021; Alugubelli et al., 2024), influenza viruses (Li et al., 2022), and HIV (Emert-Sedlak et al., 2024). BacPROTACs, which effect degradation of microbial targets through the bacterial ClpCP protease (Morreale et al., 2022), are currently being developed for the treatment of multi-drug resistant tuberculosis (Won et al., 2024; Junk et al., 2024). Preliminary reports have even suggested the utility of “TrypPROTACs” in treating trypanosomiasis and other parasitic infections (Danazumi et al., 2023; Ishii & Akiyoshi, 2022).
Since their initial description almost 25 years ago, PROTACs have proven their worth as potent and versatile therapeutic agents. Their ‘event-driven’ mechanism of action enables PROTACs to function at much lower doses than traditional drugs while retaining stringent selectivity, minimizing toxicity and off-target effects. Because PROTACs degrade rather than inhibit, they can be flexibly applied to virtually any cellular target, agnostic to enzymatic activity. Furthermore, PROTACs are less susceptible to resistance mechanisms, making them particularly well-suited for the treatment of drug-resistant cancers, antibiotic-resistant bacterial infections, and other challenging applications. Thanks to their powerful advantages, PROTAC and other TPD technologies have revolutionized the pharmaceutical landscape, and a wealth of promising preclinical data suggests that they will continue to do so in the years to come.
Sources
Alugubelli YR, Xiao J, Khatua K, Kumar S, Sun L, Ma Y, Ma XR, Vulupala VR, Atla S, Blankenship LR, Coleman D, Xie X, Neuman BW, Liu WR & Xu S. Discovery of First-in-Class PROTAC Degraders of SARS-CoV-2 Main Protease. J Med Chem 67, 6495-6507 (2024).
Arvinas, Inc. ARV-102: Arvinas’ Oral LRRK2 PROTAC Degrader. Stifel Webinar (2024).
Békés, M, Langley DR & Crews CM. PROTAC targeted protein degraders: the past is prologue. Nature Reviews Drug Discovery 21, 181-2000 (2022).
Campone M, Ma CX, De Laurentiis M, Iwata H, Hurvitz SA, Wander SA, Danso MA, Lu DR, Smith JP, Liu Y, Tran L, Anderson S & Hamilton EP. VERITAC-2: A global, randomized phase 3 study of ARV-471, a proteolysis targeting chimera (PROTAC) estrogen receptor (ER) degrader, vs fulvestrant in ER+/human epidermal growth factor receptor 2 (HER2)- advanced breast cancer. J Clinical Oncology 41, 16 (2023).
Danazumi AU, Ishmam IT, Idris S, Izert MA, Balogun EO & Górna MW. Targeted protein degradation might present a novel therapeutic approach in the fight against African trypanosomiasis. Eur Jour Pharm Sci 186, 106451 (2023).
Desantis J, Mercorelli B, Celegato M, Croci F, Bazzacco A, Baroni M, Siragusa L, Cruciani G, Loregian A & Goracci L. Indomethacin-based PROTACs as pan-coronavirus antiviral agents. Eur J Med Chem 15, 113814 (2021).
Donovan KA, Ferguson FM, Bushman JW, Eleuteri NA, Bhunia D, Ryu S, Tan L, Shi K, Yue H, Liu X, Dobrovolsky D, Jiang B, Wang J, Hao M, You I, Teng M, Liang Y, Hatcher J, Li Z, Manz TD, Groendyke B, Hu W, Nam Y, Sengupta S, Cho H, Shin I, Agius MP, Ghobrial IM, Ma MW, Che J, Buhrlage SJ, Sim T, Gray NS & Fischer ES. Mapping the Degradable Kinome Provides a Resource for Expedited Degrader Development. Cell 183, 1714-1731 (2020).
Emert-Sedlak LA, Tice CM, Shi H, Alvarado JJ, Shu ST, Reitz AB & Smithgall TE. PROTAC-mediated degradation of HIV-1 Nef efficiently restores cell-surface CD4 and MHC-I expression and blocks HIV-1 replication. Cell Chem Biol 31, 658-668 (2024).
Espinoza-Chávez RM, Salerno A, Liuzzi A, Ilari A, Milelli A, Uliassi E & Bolognesi ML. Targeted Protein Degradation for Infection Diseases: From Basic Biology to Drug Discovery. ACS BioMed Chem Au 3, 32-45 (2023).
Fang Y, Wang S, Han S, Zhao Y, Yu C, Liu H & Li N. Targeted protein degrader development for cancer: advances, challenges, and opportunities. Trends in Pharmacological Sciences 44, 303-317 (2023).
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 Clinical Oncology 40, 6 (2022)
Hamilton EP, Schott AF, Nanda R, Lu H, Keung CF, Gedrich R, Parameswaran J, Han HS & Hurvitz SA. ARV-471, an estrogen receptor (ER) PROTAC degrader, combined with palbociclib in advanced ER+/human epidermal growth factor receptor 2-negative (HER2-) breast cancer: Phase 1b cohort (part C) of a phase 1/2 study. J Clinical Oncology 40, 16 (2022).
Hatzivassiliou G, Song K, Yen I, Brandhuber BJ, Anderson DJ, Alvarado R, Ludlam MJC, Stokoe D, Gloor SL, Vigers G, Morales T, Aliagas I, Liu B, Sideris S, Hoeflich KP, Jaiswal BS, Seshagiri S, Koeppen H, Belvin M, Friedman LS & Malek S. RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature 464, 431-5 (2010).
Ishii M & Akiyoshi B. Targeted protein degradation using deGradFP in Trypanosoma brucei. Wellcome Open Res 7, 175 (2022).
Jia X & Han X. Targeting androgen receptor degradation with PROTACs from bench to bedside. Biomedicine & Pharmacotherapy 158, 114112 (2023).
Junk L, Schmiedel VM, Guha S, Fischel K, Greb P, Vill K, Krisilia V, van Geelen L, Rumpel K, Kaur P, Krishnamurthy RV, Narayanan S, Shandil RK, Singh M, Kofink C, Mantoulidis A, Biber P, Gmaschitz G, Kazmaier U, Meinhart A, Leodolter J, Hoi D, Junker S, Morreale FE, Clausen T, Kalscheuer R, Weinstabl H & Boehmelt, G. Homo-BacPROTAC-induced degradation of ClpC1 as a strategy against drug-resistant mycobacteria. Nature Commun 15, 2005 (2024).
Kim C, Wang XD, Lu Z, Zha S & Yu Y. Targeting Scaffolding Functions of Enzymes Using PROTAC Approaches. Biochemistry 62, 561-563 (2023).
Li H, Wang S, Ma W, Cheng B, Yi Y, Ma X, Xiao S, Zhang L & Zhou D. Discovery of Pentacyclic Triterpenoid PROTACs as a Class of Effective Hemagglutinin Protein Degraders. J Med Chem 65, 7154-69 (2022).
Li Z, Wang C, Wang Z, Zhu C, Li J, Sha T, Ma L, Gao C, Yang Y, Sun Y, Wang J, Sun X, Lu C, Difiglia M, Mei Y, Ding C, Luo S, Dang Y, Ding Y, Fei Y & Lu B. Allele-selective lowering of mutant HTT protein by HTT-LC3 linker compounds. Nature 575, 203-9 (2019).
Lu M, Liu T, Jiao Q, Ji J, Tao M, Liu Y, You Q & Jiang Z. Discovery of a Keap1-dependent peptide PROTAC to knockdown Tau by ubiquitination-proteasome degradation pathway. Eur J Med Chem 146, 251-9 (2018).
Lu X, Smaill JB & Ding K. Medicinal Chemistry Strategies for the Development of Kinase Inhibitors Targeting Point Mutations. J Med Chem 63, 10726-10741 (2020).
Mato A, Danilov AV, Patel MR, Tees MT, Flinn IW, Ai WZ, Patel K, Wang M, O’Brien SM, Nandakumar S, Tan M, Meredith E, Gessner M, Kim SY, Wiestner A & Wierda WG. A first-in-human phase 1 trial of NX-2127, a first-in-class oral BTK degrader with IMiD-like activity, in patients with relapsed and refractory B-cell malignancies. J Clinical Oncology 40, 16 (2022).
Morreale FE, Kleine S, Leodolter J, Junker S, Hoi DM, Ovchinnikov S, Okun A, Kley J, Kurzbauer R, Junk L, Guha S, Podlesainski D, Kazmaier U, Boehmelt G, Weinstabl H, Rumpel K, Schmiedel VM, Hartl M, Haselbach D, Meinhart A, Kaiser M & Clausen T. BacPROTACs mediate targeted protein degradation in bacteria. Cell 185, 2338-53 (2022).
Olson CM, Jiang B, Erb MA, Liang Y, Doctor ZM, Zhang Z, Zhang T, Kwiatkowski N, Boukhali M, Green JL, Haas W, Nomanbhov T, Fischer ES, Young JA, Bradner JE, Winter GE & Gray NS. Pharmacological perturbation of CDK9 using selective CDK9 inhibition or degradation. Nature Chemical Biology 14, 163-170 (2018).
Silva MC, Nandi G, Donovan KA, Cai Q, Berry BC, Nowak RP, Fischer ES, Gray NS, Ferguson FM & Haggarty SJ. Discovery and Optimization of Tau Targeted Protein Degraders Enabled by Patient Induced Pluripotent Stem Cells-Derived Neuronal Models of Tauopathy. Front Cell Neurosci 16 (2022).
Starodub A, Gollerkeri A, De savi S, Dey J, Agarwal S, Donohue S, Perea R, Klaus C & Gollob J. Phase 1 study of KT-333, a targeted protein degrader, in patients with relapsed or refractory lymphomas, large granular lymphocytic leukemia, and solid tumors.J Clinical Oncology 40, 16 (2022).
Sun, Y, Ding N, Song Y, Yang Z, Liu W, Zhu J & Rao Y. Degradation of Bruton’s tyrosine kinase mutants by PROTACs for potential treatment of ibrutinib-resistant non-Hodgkin lymphomas. Lymphoma 33, 2105-2110 (2019).
Tong Y, Zhu W, Chen J, Wen T, Xu F & Pang J. Discovery of Small-Molecule Degraders for Alpha-Synuclein Aggregates. J Med Chem 66, 7926-42 (2023).
Tsai JM, Nowak RP, Ebert BL & Fischer ES. Targeted protein degradation: from mechanisms to clinic. Nature Reviews Molecular Cell Biology (2024).
Tseng Y, Lu P, Lee C, He R, Huang Y, Tseng Y, Cheng TR, Huang JJ & Fang J. Degradation of neurodegenerative disease-associated TDP-43 aggregates and oligomers via a proteolysis-targeting chimera. J Biomed Sci 30, 27 (2023).
Wang S, He F, Tian C & Sun A. From PROTAC to TPD: Advances and Opportunities in Targeted Protein Degradation.Pharmaceuticals (Basel) 17, 100 (2024).
de Whispalaere M, Du G, Donovan KA, Zhang T, Eleuteri NA, Yuan JC, Kalabathula J, Nowak RP, Fischer ES, Gray NS & Yang PL. Small molecule degraders of the hepatitis C virus protease reduce susceptibility to resistance mutations. Nature Communication 10, 2468 (2019).
Won HI, Zinga S, Kandror O, Akopian T, Wolf ID, Schweber JTP, Schmid EW, Chao MC, Waldor M, Rubin EJ & Zhu J. Targeted protein degradation in mycobacteria uncovers antibacterial effects and potentiates antibiotic efficacy. Nat Commun 15, 4065 (2024).
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 therapeutic agents. Molecular Diversity 28, 309-333 (2024).
Zhu W, Zhang W, Chen J, Tong Y, Xu F & Pang J. Discovery of Effective Dual PROTAC Degraders for Neurodegenerative Disease-Associated Aggregates. J Med Chem 67, 3448-66 (2024).