Abstract
Heterobifunctional protein degraders, such as PROteolysis TArgeting Chimera (PROTAC) protein degraders, constitute a novel therapeutic modality that harnesses the cell’s natural protein-degradation machinery — that is, the ubiquitin–proteasome system — to selectively target proteins involved in disease pathogenesis for elimination. Protein degraders have several potential advantages over small-molecule inhibitors that have traditionally been used for cancer treatment, including their event-driven (rather than occupancy-driven) pharmacology, which permits sub-stoichiometric drug concentrations for activity, their capacity to act iteratively and target multiple copies of a protein of interest, and their potential to target nonenzymatic proteins that were previously considered ‘undruggable’. Following numerous innovations in protein degrader design and rigorous evaluation in preclinical models, protein degraders entered clinical testing in 2019. Currently, 18 protein degraders are in phase I or phase I/II clinical trials that involve patients with various tumour types, with a phase III trial of one initiated in 2022. The first safety, efficacy and pharmacokinetic data from these studies are now materializing and, although considerably more evidence is needed, protein degraders are showing promising activity as cancer therapies. Herein, we review advances in protein degrader development, the preclinical research that supported their entry into clinical studies, the available data for protein degraders in patients and future directions for this new class of drugs.
Key points
-
The concept of harnessing the natural, intracellular protein-degradation machinery (that is, the ubiquitin–proteasome system) to eliminate disease-causing proteins was proposed more than two decades ago.
-
Since then, numerous primary papers and review articles have described the mechanistic development of protein degraders and their potential as a new therapeutic approach, including for patients with cancer.
-
As of 8 January 2023, 18 heterobifunctional protein degraders are under evaluation in clinical trials in patients with various solid tumours and haematological cancers, and the first clinical data on these molecules are now emerging.
-
Preclinical data that have been disclosed for the protein degraders currently in clinical development support their target specificity and their potency in inhibiting tumour growth compared with small-molecule inhibitors.
-
Preliminary data for protein degraders that target the androgen receptor, the oestrogen receptor and BTK have shown encouraging clinical activity in patients with prostate cancer, breast cancer and chronic lymphocytic leukaemia, respectively, and results from additional ongoing clinical studies are anticipated.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$209.00 per year
only $17.42 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
References
Savage, D. G. & Antman, K. H. Imatinib mesylate–a new oral targeted therapy. N. Engl. J. Med. 346, 683–693 (2002).
Bedard, P. L., Hyman, D. M., Davids, M. S. & Siu, L. L. Small molecules, big impact: 20 years of targeted therapy in oncology. Lancet 395, 1078–1088 (2020).
Zhong, L. et al. Small molecules in targeted cancer therapy: advances, challenges, and future perspectives. Signal Transduct. Target. Ther. 6, 201 (2021).
Neklesa, T. K., Winkler, J. D. & Crews, C. M. Targeted protein degradation by PROTACs. Pharmacol. Ther. 174, 138–144 (2017).
Cromm, P. M. & Crews, C. M. Targeted protein degradation: from chemical biology to drug discovery. Cell Chem. Biol. 24, 1181–1190 (2017).
Mathijssen, R. H., Sparreboom, A. & Verweij, J. Determining the optimal dose in the development of anticancer agents. Nat. Rev. Clin. Oncol. 11, 272–281 (2014).
Lovly, C. M. & Shaw, A. T. Molecular pathways: resistance to kinase inhibitors and implications for therapeutic strategies. Clin. Cancer Res. 20, 2249–2256 (2014).
Rotow, J. & Bivona, T. G. Understanding and targeting resistance mechanisms in NSCLC. Nat. Rev. Cancer 17, 637–658 (2017).
Cocco, E., Scaltriti, M. & Drilon, A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat. Rev. Clin. Oncol. 15, 731–747 (2018).
Gebru, M. T. & Wang, H. G. Therapeutic targeting of FLT3 and associated drug resistance in acute myeloid leukemia. J. Hematol. Oncol. 13, 155 (2020).
Braun, T. P., Eide, C. A. & Druker, B. J. Response and resistance to BCR-ABL1-targeted therapies. Cancer Cell 37, 530–542 (2020).
Stephens, D. M. & Byrd, J. C. Resistance to Bruton tyrosine kinase inhibitors: the Achilles heel of their success story in lymphoid malignancies. Blood 138, 1099–1109 (2021).
Gomatou, G. et al. Mechanisms of resistance to cyclin-dependent kinase 4/6 inhibitors. Mol. Biol. Rep. 48, 915–925 (2021).
Sakamoto, K. M. et al. Protacs: chimeric molecules that target proteins to the Skp1–Cullin-F box complex for ubiquitination and degradation. Proc. Natl Acad. Sci. USA 98, 8554–8559 (2001).
Hershko, A. & Ciechanover, A. The ubiquitin system. Annu. Rev. Biochem. 67, 425–479 (1998).
Ciechanover, A., Orian, A. & Schwartz, A. L. Ubiquitin-mediated proteolysis: biological regulation via destruction. Bioessays 22, 442–451 (2000).
Nandi, D., Tahiliani, P., Kumar, A. & Chandu, D. The ubiquitin–proteasome system. J. Biosci. 31, 137–155 (2006).
Komander, D. & Rape, M. The ubiquitin code. Annu. Rev. Biochem. 81, 203–229 (2012).
Kleiger, G. & Mayor, T. Perilous journey: a tour of the ubiquitin–proteasome system. Trends Cell Biol. 24, 352–359 (2014).
Alabi, S. Novel mechanisms of molecular glue-induced protein degradation. Biochemistry 60, 2371–2373 (2021).
Frere, G. A., de Araujo, E. D. & Gunning, P. T. Emerging mechanisms of targeted protein degradation by molecular glues. Methods Cell Biol. 169, 1–26 (2022).
Hua, L. et al. Beyond proteolysis-targeting chimeric molecules: designing heterobifunctional molecules based on functional effectors. J. Med. Chem. 65, 8091–8112 (2022).
Gadd, M. S. et al. Structural basis of PROTAC cooperative recognition for selective protein degradation. Nat. Chem. Biol. 13, 514–521 (2017).
Bondeson, D. P. et al. Lessons in PROTAC design from selective degradation with a promiscuous warhead. Cell Chem. Biol. 25, 78–87 e75 (2018).
Douglass, E. F. Jr, Miller, C. J., Sparer, G., Shapiro, H. & Spiegel, D. A. A comprehensive mathematical model for three-body binding equilibria. J. Am. Chem. Soc. 135, 6092–6099 (2013).
Buckley, D. L. et al. HaloPROTACS: use of small molecule PROTACs to induce degradation of halotag fusion proteins. ACS Chem. Biol. 10, 1831–1837 (2015).
Riching, K. M., Caine, E. A., Urh, M. & Daniels, D. L. The importance of cellular degradation kinetics for understanding mechanisms in targeted protein degradation. Chem. Soc. Rev. 51, 6210–6221 (2022).
Schneekloth, A. R., Pucheault, M., Tae, H. S. & Crews, C. M. Targeted intracellular protein degradation induced by a small molecule: en route to chemical proteomics. Bioorg. Med. Chem. Lett. 18, 5904–5908 (2008).
Bekes, M., Langley, D. R. & Crews, C. M. PROTAC targeted protein degraders: the past is prologue. Nat. Rev. Drug. Discov. 21, 181–200 (2022).
Bond, M. J., Chu, L., Nalawansha, D. A., Li, K. & Crews, C. M. Targeted degradation of oncogenic KRAS(G12C) by VHL-recruiting PROTACs. ACS Cent. Sci. 6, 1367–1375 (2020).
Zhou, H. et al. SD-91 as a potent and selective STAT3 degrader capable of achieving complete and long-lasting tumor regression. ACS Med. Chem. Lett. 12, 996–1004 (2021).
Burslem, G. M. et al. Targeting BCR-ABL1 in chronic myeloid leukemia by PROTAC-mediated targeted protein degradation. Cancer Res. 79, 4744–4753 (2019).
Buhimschi, A. D. et al. Targeting the C481S ibrutinib-resistance mutation in Bruton’s tyrosine kinase using PROTAC-mediated degradation. Biochemistry 57, 3564–3575 (2018).
Azad, A. A. et al. Androgen receptor gene aberrations in circulating cell-free DNA: biomarkers of therapeutic resistance in castration-resistant prostate cancer. Clin. Cancer Res. 21, 2315–2324 (2015).
Robinson, D. et al. Integrative clinical genomics of advanced prostate cancer. Cell 161, 1215–1228 (2015).
Quigley, D. A. et al. Genomic hallmarks and structural variation in metastatic prostate cancer. Cell 174, 758–769 e759 (2018).
Takeda, D. Y. et al. A somatically acquired enhancer of the androgen receptor is a noncoding driver in advanced prostate cancer. Cell 174, 422–432 e413 (2018).
Gao, X. et al. Phase 1/2 study of ARV-110, an androgen receptor (AR) PROTAC degrader, in metastatic castration-resistant prostate cancer (mCRPC) [Abstr.]. J. Clin. Oncol. 40 (Suppl. 6), 17 (2022).
Neklesa, T. et al. ARV-110: an oral androgen receptor PROTAC degrader for prostate cancer [Abstr.]. J. Clin. Oncol. 37 (Suppl. 7S), 259 (2019).
Petrylak, D. P. et al. First-in-human phase 1 study of ARV-110, an androgen receptor (AR) PROTAC degrader in patients with metastatic castration-resistant prostate cancer following enzalutamide (ENZ) and/or abiraterone (ABI) [Abstr.]. J. Clin. Oncol. 38 (Suppl. 15), 3500 (2020).
Snyder, L. B. et al. Discovery of ARV-110, a first in class androgen receptor degrading PROTAC for the treatment of men with metastatic castration resistant prostate cancer [Abstr.]. Cancer Res. 81 (Suppl. 13), 43 (2021).
Jiang, B. et al. Development of dual and selective degraders of cyclin-dependent kinases 4 and 6. Angew. Chem. Int. Ed. Engl. 58, 6321–6326 (2019).
Nowak, R. P. et al. Plasticity in binding confers selectivity in ligand-induced protein degradation. Nat. Chem. Biol. 14, 706–714 (2018).
Degorce, S. L. et al. Discovery of proteolysis-targeting chimera molecules that selectively degrade the IRAK3 pseudokinase. J. Med. Chem. 63, 10460–10473 (2020).
Fang, Y. et al. Progress and challenges in targeted protein degradation for neurodegenerative disease therapy. J. Med. Chem. 65, 11454–11477 (2022).
Itoh, Y., Ishikawa, M., Naito, M. & Hashimoto, Y. Protein knockdown using methyl bestatin-ligand hybrid molecules: design and synthesis of inducers of ubiquitination-mediated degradation of cellular retinoic acid-binding proteins. J. Am. Chem. Soc. 132, 5820–5826 (2010).
Bondeson, D. P. et al. Catalytic in vivo protein knockdown by small-molecule PROTACs. Nat. Chem. Biol. 11, 611–617 (2015).
Zengerle, M., Chan, K. H. & Ciulli, A. Selective small molecule induced degradation of the BET bromodomain protein BRD4. ACS Chem. Biol. 10, 1770–1777 (2015).
Buckley, D. L. et al. Small-molecule inhibitors of the interaction between the E3 ligase VHL and HIF1alpha. Angew. Chem. Int. Ed. Engl. 51, 11463–11467 (2012).
Galdeano, C. et al. Structure-guided design and optimization of small molecules targeting the protein-protein interaction between the von Hippel-Lindau (VHL) E3 ubiquitin ligase and the hypoxia inducible factor (HIF) alpha subunit with in vitro nanomolar affinities. J. Med. Chem. 57, 8657–8663 (2014).
Winter, G. E. et al. Phthalimide conjugation as a strategy for in vivo target protein degradation. Science 348, 1376–1381 (2015).
Lu, J. et al. Hijacking the E3 ubiquitin ligase cereblon to efficiently target BRD4. Chem. Biol. 22, 755–763 (2015).
Ito, T. et al. Identification of a primary target of thalidomide teratogenicity. Science 327, 1345–1350 (2010).
Khan, S. et al. A selective BCL-XL PROTAC degrader achieves safe and potent antitumor activity. Nat. Med. 25, 1938–1947 (2019).
Ishida, T. & Ciulli, A. E3 ligase ligands for PROTACs: how they were found and how to discover new ones. SLAS Discov. 26, 484–502 (2021).
Chang, L., Ruiz, P., Ito, T. & Sellers, W. R. Targeting pan-essential genes in cancer: challenges and opportunities. Cancer Cell 39, 466–479 (2021).
Belcher, B. P., Ward, C. C. & Nomura, D. K. Ligandability of E3 ligases for targeted protein degradation applications. Biochemistry https://doi.org/10.1021/acs.biochem.1c00464 (2021).
Goodnow, R. A. Jr, Dumelin, C. E. & Keefe, A. D. DNA-encoded chemistry: enabling the deeper sampling of chemical space. Nat. Rev. Drug. Discov. 16, 131–147 (2017).
Disch, J. S. et al. Bispecific estrogen receptor alpha degraders incorporating novel binders identified using DNA-encoded chemical library screening. J. Med. Chem. 64, 5049–5066 (2021).
Chana, C. K. et al. Discovery and structural characterization of small molecule binders of the human CTLH E3 ligase subunit GID4. J. Med. Chem. 65, 12725–12746 (2022).
Mason, J. W. et al. DNA-encoded library (DEL)-enabled discovery of proximity-inducing small molecule. Preprint at https://doi.org/10.1101/2022.10.13.512184 (2022).
Bemis, T. A., La Clair, J. J. & Burkart, M. D. Unraveling the role of linker design in proteolysis targeting chimeras. J. Med. Chem. 64, 8042–8052 (2021).
Farnaby, W. et al. BAF complex vulnerabilities in cancer demonstrated via structure-based PROTAC design. Nat. Chem. Biol. 15, 672–680 (2019).
Xiang, W. et al. Discovery of ARD-2585 as an exceptionally potent and orally active PROTAC degrader of androgen receptor for the treatment of advanced prostate cancer. J. Med. Chem. 64, 13487–13509 (2021).
Snyder, L. B. et al. The discovery of ARV-471, an orally bioavailable estrogen receptor degrading PROTAC for the treatment of patients with breast cancer [Abstr.]. Cancer Res. 81 (Suppl. 13), 44 (2021).
Smith, B. E. et al. Differential PROTAC substrate specificity dictated by orientation of recruited E3 ligase. Nat. Commun. 10, 131 (2019).
Bricelj, A. et al. Influence of linker attachment points on the stability and neosubstrate degradation of cereblon ligands. ACS Med. Chem. Lett. 12, 1733–1738 (2021).
Schiemer, J. et al. Snapshots and ensembles of BTK and cIAP1 protein degrader ternary complexes. Nat. Chem. Biol. 17, 152–160 (2021).
Zaidman, D., Prilusky, J. & London, N. PRosettaC: Rosetta based modeling of PROTAC mediated ternary complexes. J. Chem. Inf. Model. 60, 4894–4903 (2020).
Bai, N. et al. Rationalizing PROTAC-mediated ternary complex formation using Rosetta. J. Chem. Inf. Model. 61, 1368–1382 (2021).
Weng, G., Li, D., Kang, Y. & Hou, T. Integrative modeling of PROTAC-mediated ternary complexes. J. Med. Chem. 64, 16271–16281 (2021).
Drummond, M. L., Henry, A., Li, H. & Williams, C. I. Improved accuracy for modeling PROTAC-mediated ternary complex formation and targeted protein degradation via new in silico methodologies. J. Chem. Inf. Model. 60, 5234–5254 (2020).
Poongavanam, V., Doak, B. C. & Kihlberg, J. Opportunities and guidelines for discovery of orally absorbed drugs in beyond rule of 5 space. Curr. Opin. Chem. Biol. 44, 23–29 (2018).
Lipinski, C. A., Lombardo, F., Dominy, B. W. & Feeney, P. J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug. Deliv. Rev. 46, 3–26 (2001).
DeGoey, D. A., Chen, H. J., Cox, P. B. & Wendt, M. D. Beyond the rule of 5: lessons learned from AbbVie’s drugs and compound collection. J. Med. Chem. 61, 2636–2651 (2018).
Edmondson, S. D., Yang, B. & Fallan, C. Proteolysis targeting chimeras (PROTACs) in ‘beyond rule-of-five’ chemical space: recent progress and future challenges. Bioorg. Med. Chem. Lett. 29, 1555–1564 (2019).
Pike, A., Williamson, B., Harlfinger, S., Martin, S. & McGinnity, D. F. Optimising proteolysis-targeting chimeras (PROTACs) for oral drug delivery: a drug metabolism and pharmacokinetics perspective. Drug. Discov. Today 25, 1793–1800 (2020).
Han, X. et al. Strategies toward discovery of potent and orally bioavailable proteolysis targeting chimera degraders of androgen receptor for the treatment of prostate cancer. J. Med. Chem. 64, 12831–12854 (2021).
Goracci, L. et al. Understanding the metabolism of proteolysis targeting chimeras (PROTACs): the next step toward pharmaceutical applications. J. Med. Chem. 63, 11615–11638 (2020).
Mares, A. et al. Extended pharmacodynamic responses observed upon PROTAC-mediated degradation of RIPK2. Commun. Biol. 3, 140 (2020).
Miah, A. H. et al. Optimization of a series of RIPK2 PROTACs. J. Med. Chem. 64, 12978–13003 (2021).
Bartlett, D. W. & Gilbert, A. M. A kinetic proofreading model for bispecific protein degraders. J. Pharmacokinet. Pharmacodyn. 48, 149–163 (2021).
Bartlett, D. W. & Gilbert, A. M. Translational PK-PD for targeted protein degradation. Chem. Soc. Rev. 51, 3477–3486 (2022).
Sayegh, N., Swami, U. & Agarwal, N. Recent advances in the management of metastatic prostate cancer. JCO Oncol. Pract. 18, 45–55 (2022).
Shore, N. D. et al. Phase 1b study of bavdegalutamide, an androgen receptor PROTAC degrader, combined with abiraterone in patients with metastatic prostate cancer [Abstr.]. J. Clin. Oncol. 40 (Suppl. 16), TPS5106 (2022).
Burstein, H. J. Systemic therapy for estrogen receptor-positive, HER2-negative breast cancer. N. Engl. J. Med. 383, 2557–2570 (2020).
Hanker, A. B., Sudhan, D. R. & Arteaga, C. L. Overcoming endocrine resistance in breast cancer. Cancer Cell 37, 496–513 (2020).
Nathan, M. R. & Schmid, P. A review of fulvestrant in breast cancer. Oncol. Ther. 5, 17–29 (2017).
Robertson, J. F. et al. A randomized trial to assess the biological activity of short-term (pre-surgical) fulvestrant 500 mg plus anastrozole versus fulvestrant 500 mg alone or anastrozole alone on primary breast cancer. Breast Cancer Res. 15, R18 (2013).
Kuter, I. et al. Dose-dependent change in biomarkers during neoadjuvant endocrine therapy with fulvestrant: results from NEWEST, a randomized phase II study. Breast Cancer Res. Treat. 133, 237–246 (2012).
Toy, W. et al. Activating ESR1 mutations differentially affect the efficacy of ER antagonists. Cancer Discov. 7, 277–287 (2017).
Flanagan, J. J. et al. ARV-471, an oral estrogen receptor PROTAC degrader for breast cancer. Cancer Res. 79 (Suppl. 4), P5-04-18 (2019).
Hamilton, E. P. et al. ARV-471, an estrogen receptor (ER) PROTAC degrader, combined with palbociclib in advanced ER+/human epidermal growth factor receptor 2 negative breast cancer: phase 1b cohort (part C) of a phase 1/2 study [Abstr.]. J. Clin. Oncol. 40 (Suppl. 16), TPS1120 (2022).
He, W. et al. Novel chimeric small molecule AC682 potently degrades estrogen receptor with oral anti-tumor efficacy superior to fulvestrant [Abstr.]. Cancer Res. 81 (Suppl. 4), PS18-09 (2021).
Tasso, B., Sparallarossa, A., Russo, E. & Brullo, C. The development of BTK inhibitors: a five-year update. Molecules 26, 7411 (2021).
Robbins, D. W. et al. NX-2127, a degrader of BTK and IMiD neosubstrates, for the treatment of B-cell malignancies [Abstr.]. Blood 136 (Suppl. 1), 34 (2020).
Noviski, M. et al. Concurrent degradation of BTK and IMiD neosubstrates by NX-2127 enhances multiple mechanisms of tumor killing. Cancer Res. 82 (Suppl. 12), 1128 (2022).
Montoya, S. et al. Kinase dead BTK mutations confer resistance to covalent and noncovalent BTK inhibitors but are susceptible to clinical stage BTK degraders. Blood 140 (Suppl. 1), 1811–1813 (2022).
Robbins, D. W. et al. NX-5948, a selective degrader of BTK with activity in preclinical models of hematologic and brain malignancies. Blood 138 (Suppl. 1), 2251 (2021).
Brien, G. L. et al. Targeted degradation of BRD9 reverses oncogenic gene expression in synovial sarcoma. eLife 7, e41305 (2018).
Jackson, K. L. et al. The discovery and characterization of CFT8634: a potent and selective degrader of BRD9 for the treatment of SMARCB1 perturbed cancers [Abstr.]. Cancer Res. 82 (Suppl. 12), ND09 (2022).
Collins, M. et al. Preclinical validation of target engagement assays and investigation of mechanistic impacts of FHD-609, a clinical-stage BRD9 degrader being developed for the treatment of synovial sarcoma. Presented at the 2022 Connective Tissue Oncology Society Annual Meeting. P187 (2022).
He, Y. et al. DT2216 — a Bcl-xL-specific degrader is highly active against Bcl-xL-dependent T cell lymphomas. J. Hematol. Oncol. 13, 95 (2020).
Kolb, R. et al. Proteolysis-targeting chimera against BCL-XL destroys tumor-infiltrating regulatory T cells. Nat. Commun. 12, 1281 (2021).
Togashi, Y., Shitara, K. & Nishikawa, H. Regulatory T cells in cancer immunosuppression — implications for anticancer therapy. Nat. Rev. Clin. Oncol. 16, 356–371 (2019).
Jaiswal, A. et al. Resistance to the BCL-XL degrader DT2216 in T-cell acute lymphoblastic leukemia is rare and correlates with decreased BCL-XL proteolysis. Cancer Chemother. Pharmacol. 91, 89–95 (2023).
Walker, D. et al. KTX-120, a novel IRAKIMiD degrader of IRAK4 and IMiD substrates, shows preferential activity and induces regressions in MYD88-mutant DLBCL cell and patient derived xenograft models. Blood 136 (Suppl. 1), 41 (2020).
Klaus, C. et al. Mechanisms underlying synergistic activity in MYD88MTDLBCL of KT-413, a targeted degrader of IRAK4 and IMiD substrate [Abstr.]. Cancer Res. 81 (Suppl. 13), LB118 (2021).
Liu, P. C. C. et al. A first-in-class STAT3 degrader KT-333 in development for treatment of hematologic cancers. Blood 138 (Suppl. 1), 1865 (2021).
Prior, I. A., Lewis, P. D. & Mattos, C. A comprehensive survey of Ras mutations in cancer. Cancer Res. 72, 2457–2467 (2012).
Nagashima, T. et al. ASP3082, a First-in-class novel KRAS G12D degrader, exhibits remarkable anti-tumor activity in KRAS G12D mutated cancer models. Eur. J. Cancer 174, S30 (2022).
Poulikakos, P. I., Sullivan, R. J. & Yaeger, R. Molecular pathways and mechanisms of BRAF in cancer therapy. Clin. Cancer Res. 28, 4618–4628 (2022).
Sowa, M. E. et al. Preclinical evaluation of CFT1946 as a selective degrader of mutant BRAF for the treatment of BRAF driven cancers [Abstr.]. Cancer Res. 82 (Suppl. 12), 2158 (2022).
Starodub, A. N. et al. Phase 1 study of KT-333, a targeted protein degrader of STAT3, in patients with relapsed or refractory lymphomas, large granular lymphocytic leukemia, and solid tumors [Abstr.]. J. Clin. Oncol. 40 (Suppl. 16), TPS3171 (2022).
Stevens, D. A. et al. Phase 1 study of KT-413, a targeted protein degrader, in adult patients with relapsed or refractory B-cell non-Hodgkin lymphoma. J. Clin. Oncol. 40 (Suppl. 16), TPS3170 (2022).
Mato, A. et al. 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 [Poster P649]. HemaSphere 6, 547–548 (2022).
Mato, A. et al. 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 [Abstr.]. J. Clin. Oncol. 40 (Suppl. 16), TPS7581 (2022).
Linton, K. et al. A first-in-human phase 1 trial of NX-5948, an oral BTK degrader, in patients with relapsed and refractory B-cell malignancies [Poster P650]. HemaSphere 6, 548–549 (2022).
Romanel, A. et al. Plasma AR and abiraterone-resistant prostate cancer. Sci. Transl. Med. 7, 312re310 (2015).
Lallous, N. et al. Functional analysis of androgen receptor mutations that confer anti-androgen resistance identified in circulating cell-free DNA from prostate cancer patients. Genome Biol. 17, 10 (2016).
Wyatt, A. W. et al. Genomic alterations in cell-free DNA and enzalutamide resistance in castration-resistant prostate cancer. JAMA Oncol. 2, 1598–1606 (2016).
Jernberg, E., Bergh, A. & Wikstrom, P. Clinical relevance of androgen receptor alterations in prostate cancer. Endocr. Connect. 6, R146–R161 (2017).
Hamilton, E. et al. First-in-human safety and activity of ARV-471, a novel PROTAC® estrogen receptor degrader, in ER+/HER2− locally advanced or metastatic breast cancer [Abstr.]. Cancer Res. 82 (Suppl. 4), PD13-08 (2022).
Hurvitz, S. A. et al. ARV-471, a PROTAc® estrogen receptor (ER) degrader in advanced ER-positive/human epidermal growth factor receptor 2 (HER2)-negative breast cancer: phase 2 expansion (VERITAC) of a phase 1/2 study. Abstr. GS3-03. Presented at San Antonio Breast Cancer Symposium (SABCS) (2022).
Mato, A. R. et al. NX-2127-001, a first-in-human trial of NX-2127, a Bruton’s tyrosine kinase-targeted protein degrader, in patients with relapsed or refractory chronic lymphocytic leukemia and B-cell malignancies. Blood 140 (Suppl. 1), 2329–2332 (2022).
Dragovich, P. S. Degrader-antibody conjugates. Chem. Soc. Rev. 51, 3886–3897 (2022).
Reynders, M. & Trauner, D. Optical control of targeted protein degradation. Cell Chem. Biol. 28, 969–986 (2021).
Liu, J. et al. Cancer selective target degradation by folate-caged PROTACs. J. Am. Chem. Soc. 143, 7380–7387 (2021).
Chen, H., Liu, J., Kaniskan, H. U., Wei, W. & Jin, J. Folate-guided protein degradation by immunomodulatory imide drug-based molecular glues and proteolysis targeting chimeras. J. Med. Chem. 64, 12273–12285 (2021).
Shi, S. et al. Rational design for nitroreductase (NTR)-responsive proteolysis targeting chimeras (PROTACs) selectively targeting tumor tissues. J. Med. Chem. 65, 5057–5071 (2022).
Liu, H. et al. Reactive oxygen species-responsive Pre-PROTAC for tumor-specific protein degradation. Chem. Commun. 58, 10072–10075 (2022).
Mayor-Ruiz, C. et al. Plasticity of the Cullin-RING ligase repertoire shapes sensitivity to ligand-induced protein degradation. Mol. Cell 75, 849–858 e848 (2019).
Ottis, P. et al. Cellular resistance mechanisms to targeted protein degradation converge toward impairment of the engaged ubiquitin transfer pathway. ACS Chem. Biol. 14, 2215–2223 (2019).
Zhang, L., Riley-Gillis, B., Vijay, P. & Shen, Y. Acquired resistance to BET-PROTACs (Proteolysis-Targeting Chimeras) caused by genomic alterations in core components of E3 ligase complexes. Mol. Cancer Ther. 18, 1302–1311 (2019).
Shirasaki, R. et al. Functional genomics identify distinct and overlapping genes mediating resistance to different classes of heterobifunctional degraders of oncoproteins. Cell Rep. 34, 108532 (2021).
Kurimchak, A. M. et al. The drug efflux pump MDR1 promotes intrinsic and acquired resistance to PROTACs in cancer cells. Sci. Signal. 15, eabn2707 (2022).
Acknowledgements
The authors thank J. Bloom, of Arvinas Operations, Inc. for research and editorial support. C.M.C. gratefully acknowledges NIH support (R35CA197589).
Author information
Authors and Affiliations
Contributions
K.R.H. wrote the manuscript. All authors researched data for the article, made substantial contributions to discussion of content, and reviewed/edited the manuscript before submission.
Corresponding authors
Ethics declarations
Competing interests
D.C. and K.R.H. are employees and shareholders of Arvinas Operations. C.M.C. is a founder and shareholder of Arvinas Operations, as well as a founder, shareholder and consultant of Halda Therapeutics and Siduma Therapeutics, which support research in his lab.
Peer review
Peer review information
Nature Reviews Clinical Oncology thanks A. Ciulli; M. Naito; G. Zheng, who co-reviewed with A. Smith; and the other anonymous reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Related links
ClinicalTrials.gov: https://clinicaltrials.gov/
Supplementary information
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Chirnomas, D., Hornberger, K.R. & Crews, C.M. Protein degraders enter the clinic — a new approach to cancer therapy. Nat Rev Clin Oncol 20, 265–278 (2023). https://doi.org/10.1038/s41571-023-00736-3
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41571-023-00736-3