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 11;9(1):67.
doi: 10.1038/s41698-025-00838-4.

A phase II trial of mTORC1/2 inhibition in STK11 deficient non small cell lung cancer

Affiliations

A phase II trial of mTORC1/2 inhibition in STK11 deficient non small cell lung cancer

Gary Middleton et al. NPJ Precis Oncol. .

Abstract

There are no current stratified medicine options for STK11-deficient NSCLC. STK11 loss mediates mTORC activation, GLUT1 up-regulation and increased glycolysis. This metabolic reprogramming might represent a therapeutic vulnerability targetable with mTORC1/2 inhibition. In arm B2 of the National Lung Matrix Trial 54 patients with NSCLC received vistusertib, of which 49 were STK11-deficient (30 with KRAS mutation (B2D), 19 without (B2S)). Objective response (OR) and durable clinical benefit (DCB) rates with 95% credible intervals (CrI) were estimated from posterior probability distributions generated using Bayesian beta-binomial conjugate analysis. In B2D, 2 per-protocol patients obtained OR (estimated true OR rate (95%CrI) 9.8% (2.4-24.3). Estimates of true DCB rate (95%CrI): B2D 24.4% (11.1-42.3), B2S 14.6% (3.6-34.7). Overall, vistusertib cannot be recommended in this context. Longitudinal ctDNA analysis demonstrates enrichment of SMARCA4 mutations post-treatment. In vitro studies show adaptive resistance to mTORC1/2 inhibition via AKT reactivation. (NCT02664935, ISRCTN38344105, EudraCT 2014-000814-73, 10 June 2015).

PubMed Disclaimer

Conflict of interest statement

Competing interests: G.M. received research funding from AstraZeneca and speaker bureau fees. Y.S. received an honorarium from AstraZeneca for advisory boards and educational events. A.G. and J.S.p received an honorarium from AstraZeneca for advisory boards. S.P. received consultancy and speaker bureau fees from AstraZeneca. P.J. received funding and consultancy honorarium from AstraZeneca. P.S., D.G. and D.F. received honorariums from AstraZeneca. M.B., D.M.B. and U.M. are employees of Illumina Cambridge Ltd. A.M.F. is a co-inventor on a patent application to determine methods and systems for tumour monitoring (PCT/EP2022/077987). A.D.B. received research funding from AstraZeneca. C.S. acknowledges grant support and honorarium from AstraZeneca and is an AstraZeneca advisory board member; honorarium from Illumina; is listed as an inventor on a European patent application relating to assay technology to detect tumour recurrence (PCT/GB2017/053289), the patent has been licensed to commercial entities and, under his terms of employment, C.S. is due a revenue share of any revenue generated from such license(s); holds patents relating to targeting neoantigens (PCT/EP2016/059401), identifying patient response to immune checkpoint blockade (PCT/EP2016/071471), determining HLA LOH (PCT/GB2018/052004), predicting survival rates of patients with cancer (PCT/GB2020/050221), identifying patients who respond to cancer treatment (PCT/GB2018/051912), a US patent relating to detecting tumour mutations (PCT/US2017/28013), methods for lung cancer detection (US20190106751A1) and both a European and US patent related to identifying insertion/deletion mutation targets (PCT/GB2018/051892) and is listed as a co-inventor on a patent application to determine methods and systems for tumour monitoring (PCT/EP2022/077987) and is a named inventor on a provisional patent protection related to a ctDNA detection algorithm. All other authors declare no relevant potential conflicts of interest.

Figures

Fig. 1
Fig. 1
Patient flow diagram for Arm B of the National Lung Matrix Trial.
Fig. 2
Fig. 2. Swimmer plot for participants with STK11+/- KRAS (Cohort B2) as indicated, ordered by progression-free survival time.
CT assessments are indicated and coded by Complete or Partial Response, Stable Disease, Progressive Disease or Not Evaluable according to RECIST v1.1. Follow-up time after treatment discontinuation, commencing a post-trial treatment, and death are also indicated. The solid black vertical line indicates 3 months, and the solid red line 22 weeks, being the minimum time that a participant can be counted as reaching the Durable Clinical Benefit threshold.
Fig. 3
Fig. 3. Summarised clinical endpoints (objective response rate, durable clinical benefit rate, progression-free survival, overall survival).
Forest plots indicating the median and 95% Credible Interval for (A) Objective Response rate, (B) Durable Clinical Benefit rate, (C) median Progression-free Survival time, and (D) median Overall Survival time. The vertical red lines for the co-primary outcomes of OR and DCB indicate the minimum clinically relevant threshold for the true rates that would generate a GO decision. Kaplan-Meier curves for (E) Progression-free Survival time, and (F) Overall Survival time.
Fig. 4
Fig. 4. Waterfall plot indicating the Best Percentage Change in sum of target lesion diameters for patients in the B2 cohorts.
Per-protocol participants who discontinued without having returned a measurement are included on the left of the plot with a default measurement of +100%.
Fig. 5
Fig. 5. Cell viability of STK11-mutant lung cancer cell lines treated with vistusertib (mTORC1/2 inhibition) or everolimus (mTORC1-only inhibition).
AD Cell viability of NCI-H460, A-427, NCI-A549 (STK11mut KRASmut) and ChaGoK1, NCI-H1755, CAL-12T (STK11mut KRASwt) after treatment with 0 - 10 µM vistusertib or everolimus for 48 hours, expressed as mean ± SEM from at least 3 independent experiments. Pharmacologically relevant concentrations are indicated in shaded area, calculated from previous publications,. E Relative luminescence for cells treated with 1µM everolimus vs. 1µM vistusertib (data presented from same experiements as described in AD). F shows calculated IC50 values for everolimus and vistusertib in STK11 mutant NSCLC cell lines.
Fig. 6
Fig. 6. Effect of vistusertib (AZD2014) on cell signalling.
MCF7 (control breast cancer cell line), NCI-A549 and NCI-H460 (STK11MUTKRASMUT) (left) and MCF7, ChaGo-K-1 and CAL-12T (STK11MUTKRASWT) (right) treated with 2 µM vistusertib. Lysates were collected at indicated times. Experiment performed three times on independent biological replicates, representative blots are shown.

References

    1. Cancer Genome Atlas Research Network Comprehensive molecular profiling of lung adenocarcinoma. Nature511, 543 (2014). - PMC - PubMed
    1. Sanchez-Cespedes, M. et al. Inactivation of LKB1/STK11 is a common event in adenocarcinomas of the lung. Cancer Res.62, 3659–3662 (2002). - PubMed
    1. Matsumoto, S. et al. Prevalence and specificity of LKB1 genetic alterations in lung cancers. Oncogene26, 5911–5918 (2007). - PMC - PubMed
    1. Shaw, R. J. et al. The LKB1 tumor suppressor negatively regulates mTOR signaling. Cancer Cell6, 91–99 (2004. - PubMed
    1. Gwinn, D. M. et al. AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol. Cell30, 214–226 (2008). - PMC - PubMed

LinkOut - more resources