Abstract

Abstract Multiple myeloma (MM) makes up approximately 10% of all hematologic malignancies, with increasing incidence worldwide. Though treatment options for newly diagnosed patients have evolved in recent years, they are not curative, and relapse is common with 2,500-3,000 relapsed or refractory (R/R) MM US patients seeking novel treatments, annually. Cereblon (CRBN)-binding Ikaros and Aiolos (I/A) protein degraders (i.e., lenalidomide (LEN) and pomalidomide (POM)) have had great success in treating MM. However, due to the unstable chiral nature of the CRBN-binding moiety, common to these molecules, many exist as racemic mixtures (1:1 mixture of interconverting (R)- and (S)-enantiomers). A growing body of evidence suggests that the (S)- enantiomer is the active anticancer species, while the (R)-enantiomer is inactive and potentially results in undesired activity. The ability to stabilize the active (S)- enantiomer represents a potential strategy to improve therapeutic outcomes. We are developing SP-3164, the deuterium-stabilized (S)-enantiomer of avadomide (AVA), an I/A-degrading molecular glue which has been studied in over 400 patients, demonstrating safety and activity in hematologic diseases. Our in vitro data demonstrate that SP-3164 more potently and rapidly degrades the cancer-promoting transcription factors, I/A, compared to LEN, POM, and AVA, while the stabilized (R)-enantiomer, SP-3165, neither binds CRBN nor degrades I/A. In preliminary in vivo studies, SP-3164 elicited significant tumor growth inhibition (TGI). Here, we investigate the activity of SP-3164 in MM, compared to established treatment regimens. SP-3164’s potential to degrade Aiolos in MM.1S cells was compared to SP-3165, AVA, and POM. SP-3164 rapidly and robustly degraded Aiolos in a time- and dose-dependent manner, more potently than AVA and POM, while SP-3165 demonstrated minimal Aiolos degradation. To investigate the effect of SP-3164 on cell survival, we treated NCI-H929 cells with SP-3164 (0.1 µM, 1 µM, and 10 µM) and SP-3165 (1 µM) and assessed apoptosis by flow cytometry. SP-3164 induced dose-dependent early and late apoptosis after 72 hours, while SP-3165 showed no induction of apoptosis. SP-3164’s in vivo activity was assessed and compared to SP-3165, LEN, and POM in an NCI-H929 mouse xenograft model. SP-3164 showed significant TGI as a single agent while SP-3165 seemed to support tumor growth. LEN and POM demonstrated minimal TGI. When combined with dexamethasone (DEX), SP-3164 had improved TGI compared to the combination of POM+DEX. These preclinical data demonstrate that SP-3164 is a potent CRBN-binding molecular glue with the ability to rapidly degrade cancer-promoting transcription factors and induce apoptosis in vitro. In vivo, SP-3164 has superior single agent and combination activity compared to approved molecular glues. These data support the continued development as a novel therapy in MM. Citation Format: Aundrietta D. Duncan, Daniela Y. Santiesteban, Nadeem Q. Mirza, Steve Horrigan, Sheila DeWitt, Vincent Jacques, P. Leif Bergsagel. SP-3164, a novel molecular glue degrader with activity in preclinical models of multiple myeloma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6253.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call