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
. 2019 Apr 16;27(3):806-819.e5.
doi: 10.1016/j.celrep.2019.03.066.

Pulsatile MEK Inhibition Improves Anti-tumor Immunity and T Cell Function in Murine Kras Mutant Lung Cancer

Affiliations

Pulsatile MEK Inhibition Improves Anti-tumor Immunity and T Cell Function in Murine Kras Mutant Lung Cancer

Hyejin Choi et al. Cell Rep. .

Abstract

KRAS is one of the driver oncogenes in non-small-cell lung cancer (NSCLC) but remains refractory to current modalities of targeted pathway inhibition, which include inhibiting downstream kinase MEK to circumvent KRAS activation. Here, we show that pulsatile, rather than continuous, treatment with MEK inhibitors (MEKis) maintains T cell activation and enables their proliferation. Two MEKis, selumetinib and trametinib, induce T cell activation with increased CTLA-4 expression and, to a lesser extent, PD-1 expression on T cells in vivo after cyclical pulsatile MEKi treatment. In addition, the pulsatile dosing schedule alone shows superior anti-tumor effects and delays the emergence of drug resistance. Furthermore, pulsatile MEKi treatment combined with CTLA-4 blockade prolongs survival in mice bearing tumors with mutant Kras. Our results set the foundation and show the importance of a combinatorial therapeutic strategy using pulsatile targeted therapy together with immunotherapy to optimally enhance tumor delay and promote long-term anti-tumor immunity.

PubMed Disclaimer

Conflict of interest statement

DECLARATION OF INTERESTS

P.D.S. is an employee and shareholder of AstraZeneca.

T.M. is a cofounder and holds an equity in IMVAQ Therapeutics. He is a consultant of Immunos Therapeutics and Pfizer. He has research support from Bristol-Myers Squibb; Surface Oncology; Kyn Therapeutics; Infinity Pharmaceuticals, Inc.; Peregrine Pharmeceuticals, Inc.; Adaptive Biotechnologies; Leap Therapeutics, Inc.; and Aprea. He has patents on applications related to work on oncolytic viral therapy, alpha virus-based vaccine, neo antigen modeling, CD40, GITR, OX40, PD-1, and CTLA-4.

K.K.W. is a founder and equity holder of G1 Therapeutics. He has sponsored research agreements with MedImmune, Takeda, TargImmune, and BMS. He also has consulting and sponsored research agreements with AstraZeneca, Janssen, Pfizer, Novartis, Merck, Ono, and Array.

J.D.W. is a consultant for Adaptive Biotech; Advaxis; Amgen; Apricity; Array BioPharma; Ascentage Pharma; Astellas; Bayer; Beigene; Bristol Myers Squibb; Celgene; Chugai; Elucida; Eli Lilly; F Star; Genentech; Imvaq; Janssen; Kleo Pharma; Linneaus; MedImmune; Merck; Neon Therapuetics; Ono; Polaris Pharma; Polynoma; Psioxus; Puretech; Recepta; Trieza; Sellas Life Sciences; Serametrix; Surface Oncology; and Syndax. He has research support from Bristol Myers Squibb, Medimmune, and Genentech. He holds equity in Potenza Therapeutics; Tizona Pharmaceuticals; Adaptive Biotechnologies; Elu-cida; Imvaq; Beigene; Trieza; and Linneaus and has an honorarium from Esanex. He has patents of xenogeneic DNA vaccines (royalties); alphavirus replicon particles expressing TRP2; myeloid-derived suppressor cell (MDSC) assay (royalties); Newcastle disease viruses for cancer therapy; a genomic signature to identify responders to ipilimumab in melanoma; engineered vaccinia viruses for cancer immunotherapy; an anti-CD40 agonist monoclonal antibody (mAb) fused to monophosphoryl lipid A (MPL) for cancer therapy; CAR T cells targeting differentiation antigens as means to treat cancer; an anti-PD1 antibody; anti-CTLA4 antibodies; and anti-GITR antibodies and methods of use thereof.

The other authors declare no competing interests.

Figures

Figure 1
Figure 1. MEK Inhibition Affects Murine Kras Mutant Tumor Growth and Murine T Cell Signaling
(A) pERK expression in various Kras mutant lung cancer cell lines after trametinib treatment by western blot. (B) Viability of lung tumor cell lines after selumetinib treatment. Samples were biological replicates. (C) Survival of HKP1 lung-cancer-bearing mice after 3 weeks selumetinib treatment. (D) pERK expression in CD4+ and CD8+ T cells from HKP1 tumor-bearing lungs after selumetinib or trametinib treatment by flow cytometry. Samples were biological replicates. *p < 0.05; **p < 0.01; ***p < 0.001; and ****p < 0.0001, Welch’s t test. NS, not significant. Error bars represent SD. The experiments were performed 2–3 times, and representative results are shown here.
Figure 2
Figure 2. Short Schedule of MEKi Treatment Alters T Cell Activation Status Ex Vivo
(A) Schema of ex vivo short versus long treatment experiment. (B) CTLA-4, PD1, Ki-67, and 4–1BB expression in CD8+ T cells and CD4+Foxp3 cells by flow cytometry after selumetinib (left) or trametinib (right) treatment for 96 hr. *p < 0.05; **p < 0.01, Welch’s test. Error bars represent SD. Samples were biological replicates. The experiment was performed twice, and representative results are shown here.
Figure 3
Figure 3. Short Treatment of MEKis Alters T Cell Priming Ex Vivo
(A) Schema of short treatment on Pmel-1 CD8+ T cells with human gp100 peptide pulse. (B) Flow cytometry plots of CD44 and CD62L markers on CD8+ T cells after 5 days of priming. (C) Frequency of CD44 CD62L subsets from CD8+ T cells by flow cytometry analysis. Average percentage of each subset is presented. (D) Frequency of CD44+ CD62L cell population by flow cytometry. (E) IFNγ production from supernatant at day 5 by cytokine profiling. *p < 0.05; **p < 0.01; ***p < 0.001, Welch’s test. Error bars represent SD. Samples were biological replicates. The experiment was performed twice, and representative results are shown here.
Figure 4
Figure 4. Pulsatile Treatment of Selumetinib Induces CTLA-4 and PD-1 Expression In Vivo
HKP1 transplantable lung-tumor-bearing mice were treated with selumetinib (25 mg/kg, BID) as presented in (A). After 2 weeks of treatment, lungs were collected and analyzed by flow cytometry. (A) Schema of selumetinib treatment in HKP1 lung-tumor-bearing mice in vivo. (B) Frequency of CD3+ T cell subsets in lung tumors by flow cytometry. (C) Ki-67 of diverse cell populations in lung tumors by flow cytometry. (D) Scatterplots of PD-1 and CTLA-4 marker (left) and co-inhibitory marker expression from CD3+ T cell subsets of lung tumors by flow cytometry (right). Gating controls are samples without either PD-1 or CTLA-4 antibodies. *p < 0.05; **p < 0.01; ***p < 0.001, Mann-Whitney test. Samples were biological replicates. The experiment was performed 3 times, and representative results are shown here.
Figure 5
Figure 5. Pulsatile Schedule of MEKi Treatment Delays Tumor Growth In Vivo
(A) Schema of selumetinib treatment in KRASG12C mutant genetically engineered mouse model (GEMM) of lung cancer. Treatment schedule for continuous treatment (upper panel, 25 mg/kg, BID) and pulsatile treatment (lower panel, 25 mg/kg, BID). (B) Waterfall plot showing tumor volume change at indicated time points after the continuous treatment of selumetinib. (C) Waterfall plot showing tumor volume change at indicated time points after the treatment of pulsatile dosing of either vehicle (left panel) or selumetinib (right panel). (D) Representative images of immunohistochemistry (IHC) staining of pERK (left panels) and multiplicative quick scores for quantification of pERK½ staining with vehicle control, pulsatile selumetinib, or continuous selumetinib for tumor tissue samples at the end of the treatment (right panel). Scale bars, 100 mm. *p < 0.05; ****p < 0.0001. (E) Progression-free survival of KRASG12C mice treated with vehicle control, pulsatile selumetinib, or continuous selumetinib. **p < 0.01; ***p < 0.001. Samples were biological replicates. This treatment study was performed three times, and results from all mice have been combined as presented.
Figure 6
Figure 6. Co-inhibitory Signaling Was Altered Differentially by Continuous versus Pulsatile Treatment of MEKis
(A) Flow cytometry analysis of KRASG12C mutant GEMM lung-tumor-infiltrating T cell subpopulations: CD4+, CD8+, and Tregs (CD4+Foxp3+) after continuous (left) or pulsatile (right) treatment with selumetinib as presented in Figure 5A. Lung tumors were collected at the end of treatment. *p < 0.05. NS, not significant. (B) Representative flow cytometry analysis of PD-1 levels in both CD4+ and CD8+ tumor-infiltrating T cells after continuous treatment of selumetinib. (C) Quantification of inhibitory immune checkpoint molecule expression on CD4+ (upper) and CD8+ (lower) T cells after 3 weeks of continuous selumetinib treatment. *p < 0.05; **p < 0.01. (D) Quantification of inhibitory molecules within CD4+ (left) and CD8+ (right) T lymphocyte subpopulations after 3 cycles of pulsatile selumetinib treatment. **p < 0.01. Samples were biological replicates. All mice were recruited at the same time for the treatment, and results from all mice are shown here.
Figure 7
Figure 7. Pulsatile Treatment of Selumetinib with High Dosage Impacts Immune Microenvironment Differently and Enhances Survival in Combination with Anti-CTLA-4 Treatment
(A) Schema of dosing and sample collection after either high-dose (Hi; 600 mg/kg/day; left panel) or low-dose (Lo; 50 mg/kg/day; right panel) selumetinib treatment. KrasG12DTrp53fl/fl murine transplantable tumors were treated with different dosages of selumetinib. Mouse lung tumors were collected at indicated time points. Samples were biological replicates. (B) Flow cytometry analysis of different tumor-infiltrating T cell subpopulations within total infiltrating CD45+ leukocytes at indicated time points (left). PD-L1 expression levels on tumor cells (EpCAM+), myeloid cells (CD11b+), and T cells (CD4+ and CD8+) (middle); and Ki-67 expression (right). (C) Quantification of inhibitory immune checkpoint molecules expressed on CD4+ (left) and CD8+ (right) T cells. (D) Schema of selumetinib and anti-CTLA-4 treatment on LLC transplantable tumor model. (E) Survival curve from the selumetinib and anti-CTLA-4 treatment combination in immune-competent mice (C57BL/6J). (F) Survival curve from the selumetinib and anti-CTLA-4 treatment combination in immune-deficient mice (Rag1 / ). The color code is as same as in (F). (G) Survival of the pulsatile selumetinib and anti-CTLA-4 treatment group. Survival analysis was done by Log-rank (Mantel-Cox) test. * < 0.05; ** < 0.01. Samples were biological replicates. The experiment was performed 2–3 times, and representative results are shown here.

Similar articles

Cited by

References

    1. Andrzejewski S, Klimcakova E, Johnson RM, Tabariés S, Annis MG, McGuirk S, Northey JJ, Chénard V, Sriram U, Papadopoli DJ, et al. (2017). PGC-1a promotes breast cancer metastasis and confers bioenergetic flexibility against metabolic drugs. Cell Metab 26, 778–787.e5. - PubMed
    1. Bolstad BM, Irizarry RA, Astrand M, and Speed TP (2003). A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics 19, 185–193. - PubMed
    1. Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, Chow LQ, Vokes EE, Felip E, Holgado E, et al. (2015). Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N. Engl. J. Med 373, 1627–1639. - PMC - PubMed
    1. Brahmer J, Reckamp KL, Baas P, Crinò L, Eberhardt WEE, Poddubskaya E, Antonia S, Pluzanski A, Vokes EE, Holgado E, et al. (2015). Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N. Engl. J. Med 373, 123–135. - PMC - PubMed
    1. Brant R, Sharpe A, Liptrot T, Dry JR, Harrington EA, Barrett JC, Whalley N, Womack C, Smith P, and Hodgson DR (2017). Clinically viable gene expression assays with potential for predicting benefit from MEK inhibitors. Clin. Cancer Res 23, 1471–1480. - PubMed

Publication types

MeSH terms