Skip to main content

Targeted protein degradation in hematologic malignancies: latest updates from the 2023 ASH annual meeting

Abstract

Protein degraders, emerging as a novel class of therapeutic agents, have gained widespread attention due to their advantages. They have several advantages over traditional small molecule inhibitors, including high target selectivity and ability to target “undruggable” targets and overcome inhibitor drug resistance. Tremendous research and development efforts and massive investment have resulted in rapid advancement of protein degrader drug discovery in recent years. Here, we overview the latest clinical and preclinical updates on protein degraders presented at the 2023 ASH Annual Meeting.

To the editor

Small molecule inhibitors are major therapies used to treat hematologic malignancies in clinic, yet challenges such as drug resistance and off-target effects persist. Proteolysis-targeting chimeras (PROTACs) and molecular glues have emerged as promising therapeutic modality, celebrated for their ability to selectively target and efficiently degrade a wide array of proteins. This report summarizes the latest developments in protein degraders presented at 2023 ASH annual meeting.

Clinical trials

Bruton’s tyrosine kinase (BTK), crucial for B-cell receptor signaling, has been identified as a key therapeutic target in B-cell malignancies. Three preliminary clinical trials assessed the safety and efficacy of BTK PROTAC degraders. In a phase 1 trial, BGB-16673, a potent BTK degrader, showed an overall response rate of 67% (12/18), including a mantle cell lymphoma patient achieving complete response (CR) after a median follow-up of 3.5 months in relapsed or refractory B-cell malignancies. The drug showed profound and enduring reductions of BTK protein even at the lowest doses. Moreover, although with 88.5% of patients experienced treatment-emergent adverse events (TEAEs), there were no discontinuations due to adverse events [1] (Table 1). NX-5948, a novel orally administered BTK PROTAC degrader, exhibited good tolerability with no dose-limiting toxicities or serious TEAEs. Following a median treatment duration of 2.8 months, 1 out of 3 evaluated chronic lymphocytic leukemia (CLL) patient achieved a partial response (PR) [2] (Table 1). NX-2127 is an oral dual-function PROTAC degrader targeting BTK and immunomodulatory factors Ikaros and Aiolos. NX-2127 showed potent and consistent degradation of target proteins. However, there were safety concerns, as some patients discontinued their treatment due to TEAEs. After a median follow-up of 9.5 months, lasting CR were noted in non-Hodgkin’s lymphoma patients [3] (Table 1).

Table 1 Clinical trials of protein degrader presented at ASH 2023

Aberrant activation of phosphorylated signal transducer and activator of transcription 3 (STAT3) underlies various malignancies. KT-333, a STAT3 PROTAC degrader, achieved robust and dose-dependent degradation of STAT3 in a Phase 1a/1b study, with all the mean maximum degradation exceeding 60% across drug doses 1 to 4. Following two treatment cycles, one patient achieved PR. Importantly, no TEAEs were reported, suggesting a good safety profile of KT-333 [4] (Table 1).

Preclinical studies

Several degraders targeting other pathways used alone or in combination, demonstrated good efficacy in pre-clinical tumor models. Many hematological malignancies feature the overexpression of anti-apoptotic proteins, such as BCL-2 and BCL-XL. Xie et al. reported that NXD02, a novel BCL-XL PROTAC degrader, displaying superior degradation of BCL-XL (DC50 of 6.6 vs. 17.4nM) and enhanced anti-tumor efficacy (100% vs. 36%) compared to DT2216, the initial BCL-XL PROTAC degrader [5](Table 2). The BCL-2/BCL-XL-dual degraders, WH25244 and PZ18753b, demonstrated impressive efficacy against venetoclax-resistant (VR) CLL cells [6] (Table 2). Additionally, combination therapy of DT2216 with azacitidine, ruxolitinib, or S63845, revealed synergistic effects against JAK2-mutant acute myeloid leukemia (AML) [7] (Table 2).

Table 2 Preclinical studies of protein degrader presented at ASH 2023

In MYC/BCL2-related lymphoma, bromo and extra terminal (BET) inhibitors have shown limited efficacy in suppressing MYC expression. A novel BET PROTAC degrader K-256 effectively degraded bromodomain-containing protein 4 (BRD4) at low concentrations, leading to stronger tumor growth inhibition compared to existing inhibitors and other degraders [8] (Table 2).

STAT3 regulates anti-apoptotic protein MCL-1, which is implicated in VR AML. A STAT3 degrader significantly (> 60%) decreased MCL-1 and STAT3 levels, triggering apoptosis in VR AML cells. It also led to erythroid and myeloid differentiation induction of stem and progenitor cells [9] (Table 2).

Aiolos and Ikaros inhibit anti-tumor immunity through upregulating exhaustion markers in T cells, which blocking T cell activation and its tumor-killing activity. Mezigdomide, an Aiolos/Ikaros molecular glue degrader, effectively reversed the exhaustion of T cells, boosting the efficacy of immunotherapies in multiple myeloma [10] (Table 2).

Casein kinase 1α (CK1α), a multifunctional serine/threonine kinase, is involved in deactivating p53 function. PinA1, a selective CK1α molecular glue degrader, enhances p53 level and induces apoptosis in AML cells with wild-type TP53, while sparing normal cells. Additionally, PinA1 combined with FLT3, BCL-2, or MDM2 inhibitors showed enhanced anti-tumor effects in both in vitro and in vivo settings [11] (Table 2).

Molecular glue degraders such as pomalidomide and lenalidomide have demonstrated remarkable clinical success in treating hematological malignancies including multiple myeloma, and certain lymphomas. In addition to molecular glue, a lot of the recent efforts have been focused on developing PROTACs to overcome tumor resistance to inhibitor drugs. Early clinical studies and preclinical studies mentioned above have shown strong anti-tumor activities, both as standalone treatments and in combination therapies. These protein degraders showed their activities in overcoming resistance, reducing off-target effects, and enhancing treatment efficacy. Interestingly, concerns regarding the possibility of PROTACs resistance have promoted research into looking for alternative strategies, including the use of new E3 ligases other than only cereblon (CRBN) [12, 13]. While this is a very interesting concept, much more work is still needed to better define the strategy. We look forward to more comprehensive trial results that would provide further insights into the clinical utility of those novel protein degraders, and hope those degraders currently in preclinic stage will soon get into clinical trials where the therapeutic hypothesis of degrading those targets can be tested in clinic.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

ASH:

American Society of Hematology

PROTACs:

Proteolysis-targeting chimeras

BTK:

Bruton’s tyrosine kinase

CR:

Complete response

TEAEs:

Treatment-emergent adverse events

CLL:

Chronic lymphocytic leukemia

STAT3:

Signal transducer and activator of transcription 3

VR:

Venetoclax-resistant

AML:

Acute myeloid leukemia

BET:

Bromo and extra terminal

BRD4:

Bromodomain-containing protein

CK1α:

Casein kinase 1α

CRBN:

Cereblon

References

  1. Seymour JF, Cheah CY, Parrondo R, Thompson MC, Stevens DA, Lasica M, et al. First results from a phase 1, first-in-human study of the Bruton’s tyrosine kinase (BTK) degrader Bgb-16673 in patients (pts) with relapsed or refractory (R/R) B-Cell malignancies (BGB-16673-101). Blood. 2023;142(Supplement 1):4401. https://doi.org/10.1182/blood-2023-180109.

    Article  Google Scholar 

  2. Searle E, Forconi F, Linton K, Danilov A, McKay P, Lewis D, et al. Initial findings from a first-in-human phase 1a/b trial of NX-5948, a selective Bruton’s tyrosine kinase (BTK) degrader, in patients with Relapsed/Refractory B cell malignancies. Blood. 2023;142(Supplement 1):4473. https://doi.org/10.1182/blood-2023-179508.

    Article  Google Scholar 

  3. Danilov A, Tees MT, Patel K, Wierda WG, Patel M, Flinn IW, et al. A first-in-human phase 1 trial of NX-2127, a first-in-class Bruton’s tyrosine kinase (BTK) Dual-targeted protein degrader with Immunomodulatory Activity, in patients with Relapsed/Refractory B cell malignancies. Blood. 2023;142(Supplement 1):4463. https://doi.org/10.1182/blood-2023-179872.

    Article  Google Scholar 

  4. Shastri A, Feldman EJ, Starodub AN, Feldman T, Rodriguez CP, Epstein-Peterson ZD, et al. Preliminary Safety, Pharmacokinetics, Pharmacodynamics and Clinical Activity of KT-333, a targeted protein degrader of STAT3, in patients with relapsed or refractory lymphomas, large Granular lymphocytic leukemia, and solid tumors. Blood. 2023;142(Supplement 1):3081. https://doi.org/10.1182/blood-2023-181130.

    Article  Google Scholar 

  5. Xie Y, Xing W, Lai W, Tao M, Gu L, Cao M, et al. Discovery of BCL-XL Heterobifunctional Degrader with potentially improved therapeutic window and minimal platelet toxicity for hematological malignancies. Blood. 2023;142(Supplement 1):117. https://doi.org/10.1182/blood-2023-184887.

    Article  Google Scholar 

  6. Diaz Rohena D, Peris Cuesta A, Slawin B, Ravikrishnan J, Liu C, Williams C, et al. Enriched signalling pathways in Venetoclax-relapsed chronic lymphocytic leukemia (CLL) cells and targeting using a Protac-based Bcl-2/Bcl-Xl degrader. Blood. 2023;142(Supplement 1):4195. https://doi.org/10.1182/blood-2023-188250.

    Article  Google Scholar 

  7. WANG Z, Zhang Q, Skwarska A, Ramage CL, Zheng G, Gritsman K, et al. Targeting BCL-XL with a novel VHL-Based BCL-XL Degrader DT-2216 in Pre-clinical JAK2-Mutant AML Post-MPN models. Blood. 2023;142(Supplement 1):4154. https://doi.org/10.1182/blood-2023-178813.

    Article  Google Scholar 

  8. Furukawa K, Shimada K, Esaki M, Tanaka K, Yamamoto K, Mori K, et al. Development and efficacy of a Novel Bromodomain and Extraterminal Domain Degrader K-256 in MYC/BCL2-Related lymphoma. Blood. 2023;142(Supplement 1):5008. https://doi.org/10.1182/blood-2023-179334.

    Article  Google Scholar 

  9. Chakraborty S, Morganti C, Dey J, Zhang H, Aluri S, Gitego N, et al. A STAT3 degrader demonstrates pre-clinical efficacy in Venetoclax resistant Acute Myeloid Leukemia. Blood. 2023;142(Supplement 1):2787. https://doi.org/10.1182/blood-2023-186947.

    Article  Google Scholar 

  10. Chiu H, Zhao J, Ortiz Estevez M, Hagner PR, Gandhi AK. Mezigdomide reverses T-Cell exhaustion through Degradation of Aiolos/Ikaros and Reinvigoration of Cytokine Production pathways. Blood. 2023;142(Supplement 1):335. https://doi.org/10.1182/blood-2023-189445.

    Article  Google Scholar 

  11. Kim B-R, Kang H, Kim Y-J, Son H, Park J, Park Y, et al. Anti-leukemic effects of CK1α degrader as Monotherapy and in combination with targeted drugs. Blood. 2023;142(Supplement 1):2799. https://doi.org/10.1182/blood-2023-178971.

    Article  Google Scholar 

  12. Bekes M, Langley DR, Crews CM. PROTAC targeted protein degraders: the past is prologue. Nat Rev Drug Discovery. 2022;21(3):181–200. https://doi.org/10.1038/s41573-021-00371-6.

    Article  CAS  PubMed  Google Scholar 

  13. Casan JM, Seymour JF. Degraders upgraded: the rise of PROTACs in Haematological Malignancies. Blood. 2024. https://doi.org/10.1182/blood.2023022993.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by National Natural Science Foundation (No.82170189, No.81800194); Shandong Provincial Natural Science Foundation (ZR2021YQ51); Taishan Scholars Program of Shandong Province.

Author information

Authors and Affiliations

Authors

Contributions

GZ, RK and CW drafted this manuscript. GZ, SF and QH prepared tables. WX and XZ revised the manuscript. XZ provided direction and guidance throughout the preparation of the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Weilin Xie or Xiangxiang Zhou.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhong, G., Kong, R., Feng, S. et al. Targeted protein degradation in hematologic malignancies: latest updates from the 2023 ASH annual meeting. J Hematol Oncol 17, 14 (2024). https://doi.org/10.1186/s13045-024-01533-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13045-024-01533-w

Keywords