Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2025 Mar 3;15(3):578-594.
doi: 10.1158/2159-8290.CD-24-0840.

Discovery of BBO-8520, a First-In-Class Direct and Covalent Dual Inhibitor of GTP-Bound (ON) and GDP-Bound (OFF) KRASG12C

Affiliations

Discovery of BBO-8520, a First-In-Class Direct and Covalent Dual Inhibitor of GTP-Bound (ON) and GDP-Bound (OFF) KRASG12C

Anna E Maciag et al. Cancer Discov. .

Abstract

Approved inhibitors of KRASG12C prevent oncogenic activation by sequestering the inactive, GDP-bound (OFF) form rather than directly binding and inhibiting the active, GTP-bound (ON) form. This approach provides no direct target coverage of the active protein. Expectedly, adaptive resistance to KRASG12C (OFF)-only inhibitors is observed in association with increased expression and activity of KRASG12C(ON). To provide optimal KRASG12C target coverage, we have developed BBO-8520, a first-in-class, direct dual inhibitor of KRASG12C(ON) and (OFF) forms. BBO-8520 binds in the Switch-II/Helix3 pocket, covalently modifies the target cysteine, and disables effector binding to KRASG12C(ON). BBO-8520 exhibits potent signaling inhibition in growth factor-activated states, in which current (OFF)-only inhibitors demonstrate little measurable activity. In vivo, BBO-8520 demonstrates rapid target engagement and inhibition of signaling, resulting in durable tumor regression in multiple models, including those resistant to KRASG12C(OFF)-only inhibitors. BBO-8520 is in phase 1 clinical trials in patients with KRASG12C non-small cell lung cancer. Significance: BBO-8520 is a first-in-class direct, small molecule covalent dual inhibitor that engages KRASG12C in the active (ON) and inactive (OFF) conformations. BBO-8520 represents a novel mechanism of action that allows for optimal target coverage and delays the emergence of adaptive resistance seen with (OFF)-only inhibitors in the clinic. See related commentary by Zhou and Westover, p. 455.

PubMed Disclaimer

Conflict of interest statement

All BridgeBio Oncology Therapeutics (BBOT) authors are employees and stockholders of BBOT, a private company. B. Wang, R. Xiu, E. Wallace, Z. Zhang, and Y. Yang report a patent for PCT/US2022/037992(WO2023004102A2) pending. A.E. Maciag reports support from a Collaborative Research and Development Agreement with TheRas/BridgeBio and from NCI contract 75N91019D00024 during the conduct of the study, as well as a patent for PCT/US2022/037992(WO2023004102A2) pending, licensed, and with royalties paid from TheRas/BridgeBio. A.K. Sharma reports support from a Collaborative Research and Development Agreement with TheRas/BridgeBio and from NCI contract 75N91019D00024 during the conduct of the study. A.H. Chan reports support from a Collaborative Research and Development Agreement with TheRas/BridgeBio and from NCI contract 75N91019D00024 during the conduct of the study, as well as a patent for PCT/US2022/037992 (WO2023004102A2) pending, licensed, and with royalties paid from TheRas/BridgeBio. M. Dyba reports support from a Collaborative Research and Development Agreement with TheRas/BridgeBio and from NCI contract 75N91019D00024 during the conduct of the study and owning stock in BridgeBio. B.P. Smith reports support from a Collaborative Research and Development Agreement with TheRas/BridgeBio and from NCI contract 75N91019D00024 during the conduct of the study, as well as owning stock in BridgeBio Pharma, Inc. D. Rabara reports support from a Collaborative Research and Development Agreement with TheRas/BridgeBio and from NCI contract 75N91019D00024 during the conduct of the study. E.K. Larsen reports support from a Collaborative Research and Development Agreement with TheRas/BridgeBio and from NCI contract 75N91019D00024 during the conduct of the study. J.-P. Denson reports support from a Collaborative Research and Development Agreement with TheRas/BridgeBio and from NCI contract 75N91019D00024 during the conduct of the study. P.A. Alexander reports support from a Collaborative Research and Development Agreement with TheRas/BridgeBio and from NCI contract 75N91019D00024 during the conduct of the study. M. Abreu Blanco reports support from a Collaborative Research and Development Agreement with TheRas/BridgeBio and from NCI contract 75N91019D00024 during the conduct of the study. D.M. Turner reports support from a Collaborative Research and Development Agreement with TheRas/BridgeBio and from NCI contract 75N91019D00024 during the conduct of the study, as well as a patent for PCT/US2022/037992 (WO2023004102A2) pending, licensed, and with royalties paid from TheRas/BridgeBio. F.C. Lightstone reports a patent for PCT/US2022/037992(WO2023004102A2) pending and licensed to BBOT. K.-K. Wong reports grants from BridgeBio and Mirati during the conduct of the study, as well as other support from Cogent, Iambic, Pfizer, and Janssen outside the submitted work. A.G. Stephen reports support from a Collaborative Research and Development Agreement with TheRas/BridgeBio and from NCI contract 75N91019D00024 during the conduct of the study. D.K. Simanshu reports support from a Collaborative Research and Development Agreement with TheRas/BridgeBio and from NCI contract 75N91019D00024 during the conduct of the study, as well as a patent for PCT/US2022/037992 (WO2023004102A2) pending, licensed, and with royalties paid from TheRas/BridgeBio. D.V. Nissley reports support from a Collaborative Research and Development Agreement with TheRas/BridgeBio and from NCI contract 75N91019D00024 during the conduct of the study. F. McCormick reports personal fees from BBOT and Leidos Biomedical during the conduct of the study, as well as personal fees from BridgeBio, Quanta, and Amgen outside the submitted work. No disclosures were reported by the other authors.

Figures

Figure 1.
Figure 1.
Discovery of BBO-8520. A, The compound progression chart shows the development of BBO-8520. Noncovalent KRASG12D inhibitor, compound 1, showed binding activity (SPR) to GppNHp-bound KRASG12D, and disruption of KRASG12D/RAF1(RBD) interaction in PPI assay. Compound 1 was equipped with a covalent warhead to generate compound 2. Optimization of cell potency and ADME properties gave rise to compound 3. Further improvement of the GTP-bound KRASG12C activity by replacing the aminobenzothiazole of compound 3 with a cyano-amino benzothiophene at quinazoline 7-position gave rise to BBO-8520. BBO-8520 shows covalent labeling of C12 in the active, GppNHp-, or GTP-bound KRASG12C, as well as disruption of KRASG12C/RAF1(RBD) binding. B, The table summarizes the ability of compounds to modify C12 of GDP-, GppNHp-, and GTP-bound KRASG12C as measured by MALDI-TOF mass spectrometry; and their ability to disrupt GppNHp- or GTP-bound KRASG12C binding to RAF1(RBD) in PPI assay. C, kinact/Ki measurements of sotorasib, adagrasib, and BBO-8520 in the GDP-bound (OFF) and GTP-bound (ON) conformations of KRASG12C.
Figure 2.
Figure 2.
Binding mode of BBO-8520 to KRASG12C (ON) and (OFF) conformation. A, GDP-bound (OFF; top, PDB 8V3A) and GppNHp-bound (ON; bottom, PDB 8V39) forms in surface and ribbon representations. The protein is colored light orange (OFF) or light blue (ON), with Switch-I, Switch-II, and Helix 3 highlighted in pink, cyan, and orange, respectively. BBO-8520 (light green at the top, pink at the bottom), nucleotide, C12, and T35 are shown as sticks, and Mg2+ (green) and the coordinating waters (red) are shown as spheres. B and C, Enlarged view of the binding pocket in the GDP-bound form (PDB 8V3A), focusing on the regions around (B) Switch-II and Helix 3, and (C) C12 and GDP. The protein and BBO-8520 are colored light orange and light green, respectively. H-bonds are indicated by dashed lines. D and E, Enlarged view of the binding pocket in the GppNHp-bound form (PDB 8V39), focusing on the regions around (D) Switch-II and Helix 3, and (E) C12 and GppNHp. The protein and BBO-8520 are colored light blue and light pink, respectively. F and G, Overlay of the GDP-bound and GppNHp-bound structures, focusing on the regions around (F) Switch-II and Helix 3, and (G) C12 and the nucleotide.
Figure 3.
Figure 3.
Mechanism of action and selectivity of BBO-8520. A, BBO-8520 [ligand (L)] binding to KRASG12C-GTP [protein (P)] shifts the state 1–state 2 equilibrium of protein to the inactive, state 1-like conformation (induced γ1PL peak located most downfield) in the protein–ligand (PL) binary complex spectrum. Peak β1PL represents L binding induced inactive conformation of β GTP. Chemical shifts corresponding to peaks α1, β1, and γ1 belong to state 1 (inactive, effector binding–deficient) conformation, whereas α2, β2, and γ2 to the state 2 (active, effector binding–enabled) conformation. RAF1 RBD loading is unable to induce γ2 (active conformation) population (see P + L + RBD spectrum). Shown on top and bottom are the control spectra (in the presence of DMSO) of P + RBD and P, respectively. The γ1PL peak emergence is only noted in the presence of an inhibitor. The control spectra of KRASG12C-GTP alone or in the presence of RBD do not show this peak. B, BBO-8520 inhibits SOS-mediated nucleotide exchange of GDP with BODIPY-GDP KRAS. Avi-KRAS mutants indicated and Avi-NRAS WT were loaded with BODIPY-GDP, then BBO-8520 was added in a 2-fold dilution series starting at 30 nmol/L. The assay was started by the addition of SOS1 (aa564-1048) and GDP, then analyzed after 4 and 24 hours of incubation. KRASG12C shows the highest inhibition of nucleotide exchange with BBO-8520. NRAS WT was used as a control. C, pERK inhibitory activity of BBO-8520, sotorasib, and adagrasib against a panel of mouse embryonic fibroblasts (MEF) driven by KRAS mutants, HRAS, NRAS, and BRAFV600E. BBO-8520 demonstrates the best pERK inhibitory activity in KRASG12C-driven MEF cells, compared with MEFs driven by other KRAS mutants or WT KRAS. BBO-8520 shows no pERK inhibitory activity in MEFs driven by HRAS, NRAS, or BRAFV600E.
Figure 4.
Figure 4.
BBO-8520 potently inhibits KRASG12C signaling in tumor cells. A, Target engagement and ERK phosphorylation time course in the KRASG12C cancer cell lines MIA PaCa-2 and SW1473. BBO-8520 at 20 nmol/L displays rapid pERK inhibition at 30 minutes which is sustained for up to 24 hours and is compared with 20 or 100-nmol/L sotorasib and adagrasib, which take longer and show less inhibition of pERK. B, HTRF analysis of phosphorylated ERK demonstrates time- and dose-dependent inhibition in response to BBO-8520 in MIA PaCa-2 and SW1463 cells, which is sustained for up to 34 hours posttreatment. C to E, Potent effects of BBO-8520 on 2-hour pERK inhibition, pAKT inhibition, and 7-day 3D viability measurements in KRASG12C cell lines as compared with sotorasib, adagrasib, and RMC-6291. The activity of BBO-8520 in KRASG12C cell lines is compared against a cell line panel comprising KRAS wild type along with G12C/D/V and S, G13D, and BRAFV600Emutants. IC50 (nmol/L) values for each cell line are captured in Supplementary Table S2.
Figure 5.
Figure 5.
BBO-8520 maintains potency in the active state of KRASG12C. A, RAS:RAF ELISA assay in MIA PaCa-2 cells shows rapid dissociation of KRASG12C (ON) with RAF1 by BBO-8520 compared with the (OFF)-only KRASG12C inhibitors sotorasib, adagrasib, or GDC-6036. B and C, NCI-H358 cells were serum starved and then treated with 100 ng/mL of EGF, compound, and assayed for pERK HTRF 20 minutes after compound addition (B) or treated with 100 ng/mL of HGF and compound and assayed for a 5-day viability assay (C). Average potency shifts are shown to the right demonstrating the fold changes of IC50 following EGF or HGF stimulation compared with vehicle. D, HeLa cells were engineered to express a KRASG12C/A59G double mutant known for attenuated GTP hydrolysis and assayed for pERK inhibition following treatment with 0.3, 1, 3, and 10 µmol/L of BBO-8520, sotorasib, or adagrasib. BBO-8520 demonstrated a potent inhibition of the pERK signal compared with the (OFF)-only inhibitors, which showed no activity. E, A long-term clonogenic assay using IC90 concentrations (2-hour pERK) of BBO- 8520, sotorasib, and adagrasib, shows that only BBO-8520 can drive complete growth suppression for up to 35 days in culture compared with sotorasib or adagrasib.
Figure 6.
Figure 6.
BBO-8520 demonstrates dose- and time-dependent inhibition of pERK and strong efficacy in KRASG12C models. A, BBO-8520 shows dose-responsive inhibition of pERK at 6 hours following a dose of 3, 10, and 30 mg/kg in a MIA PaCa-2 Matrigel plug PD assay (*, P < 0.01; **, P < 0.0001). B, Suppression of pERK was observed up to 72 hours following treatment with 30 mg/kg of BBO-8520 in the MIA PaCa-2 Matrigel plug PD (*, P < 0.01; **, P < 0.0001). C, In the corresponding MIA PaCa-2 CDX model, BBO-8520 showed significant antitumor activity at 0.1, 0.3, 1, 3, and 10 mg/kg following 28 days of treatment (*, P < 0.0001). D, In the NCI-H358 CDX model, BBO-8520 demonstrated significant and robust efficacy at 0.3, 1, 3, and 10 mg/kg following 28 days of treatment (*, P < 0.0001). E, In the KCP NSCLC GEMM, BBO- 8520 demonstrated significant and robust efficacy at 10 mg/kg (*, P < 0.0001). F, In the CT26-KRASG12C-luciferase syngeneic liver tumor model, BBO-8520 extended the median survival as a monotherapy or in combination with anti-PD-1.
Figure 7.
Figure 7.
BBO-8520 is more efficacious than sotorasib and shows activity in sotorasib-resistant tumors. A, Efficacy of BBO-8520 and sotorasib in the RET-amplified LUN055 PDX model. Both BBO-8520 and sotorasib demonstrate antitumor activity (*, P < 0.0001), with BBO-8520 showing significant tumor regressions (23%) vs. 71% TGI for sotorasib. B, MIA PaCa-2 xenografts were grown under the presence of 10 mg/kg of sotorasib until tumors became resistant under treatment (day 35). On day 35, a cohort of eight mice were switched from sotorasib (10 mg/kg) to 30 mg/kg of BBO-8520. These mice showed strong responses with tumor volume regression. C, Analysis of sotorasib-resistant tumors showing a high proportion of KRASG12C amplification by ddPCR. D, Focused view of mice continuing on 10-mg/kg sotorasib or switched to 30-mg/kg BBO-8520 starting on day 35 (200 mm3). All mice treated with BBO-8520 had a statistically significant (P < 0.01) reduction in tumor volume compared with those continued on sotorasib. This included three mice with complete tumor regressions.

References

    1. Zehir A, Benayed R, Shah RH, Syed A, Middha S, Kim HR, et al. . Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med 2017;23:703–13. - PMC - PubMed
    1. Moore AR, Rosenberg SC, McCormick F, Malek S. RAS-targeted therapies: is the undruggable drugged? Nat Rev Drug Discov 2020;19:533–52. - PMC - PubMed
    1. Nassar AH, Adib E, Kwiatkowski DJ. Distribution of KRASG12C somatic mutations across race, sex, and cancer type. N Engl J Med 2021;384:185–7. - PubMed
    1. Lito P, Solomon M, Li L-S, Hansen R, Rosen N. Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism. Science 2016;351:604–8. - PMC - PubMed
    1. Canon J, Rex K, Saiki AY, Mohr C, Cooke K, Bagal D, et al. . The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature 2019;575:217–23. - PubMed

MeSH terms

LinkOut - more resources