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 Oct 15;85(20):3966-3982.
doi: 10.1158/0008-5472.CAN-25-0998.

TNG260 Is a Small-Molecule CoREST Inhibitor That Sensitizes STK11-Mutant Tumors to Anti-PD-1 Immunotherapy

Affiliations

TNG260 Is a Small-Molecule CoREST Inhibitor That Sensitizes STK11-Mutant Tumors to Anti-PD-1 Immunotherapy

Leanne G Ahronian et al. Cancer Res. .

Abstract

Patients with non-small cell lung cancer (NSCLC) with loss of the tumor suppressor gene STK11 are resistant to immune checkpoint therapies like anti-PD-1. In this study, we conducted an in vivo CRISPR screen that identified histone deacetylase 1 as a target to reverse anti-PD-1 resistance driven by loss of STK11 and developed TNG260, a potent small-molecule inhibitor of the CoREST complex with selectivity exceeding previously generated inhibitors in this class in preclinical studies. Treatment with TNG260 led to increased expression of immunomodulatory genes in STK11-deficient cancer cells. When combined with anti-PD-1, TNG260 induced immune-mediated stasis and/or regression in STK11-deficient syngeneic tumor models and autochthonous NSCLC models. In the tumors of patients with STK11-deficient cancers in a clinical trial (NCT05887492), treatment with a combination of TNG260 and pembrolizumab increased intratumoral histone acetylation, PD-L1 tumor proportion scores, and T-cell infiltration into the tumor microenvironment. This study illustrates a promising treatment strategy for addressing immune evasion in patients with STK11-mutant NSCLC.

Significance: Targeting CoREST with TNG260 sensitizes STK11-deficient non-small cell lung cancer to anti-PD-1 immunotherapy, offering a potential treatment for patients not served by existing therapies. See related commentary by Lin and Shen, p. 3821.

PubMed Disclaimer

Conflict of interest statement

L.G. Ahronian reports personal fees from Tango Therapeutics during the conduct of the study, as well as a patent for WO2024030659A1 issued to Tango Therapeutics. M. Zhang reports other support from Tango Therapeutics outside the submitted work. G.S. Falchook reports other support from Tango Therapeutics during the conduct of the study, as well as other support from Wolters Kluwer (royalties), other support from AbbVie (advisory role), Fujifilm (advisory role), Silicon (advisory role), Navire (advisory role), Turning Point (advisory role), Predicine (advisory role), Inspirna (advisory role), Regeneron (advisory role), Jubilant (advisory role), BostonGene (advisory role), Teon (advisory role), Merck (advisory role), Sanofi (advisory role), BridgeBio (advisory role), Avistone (advisory role), and EMD Serono (advisory role), other support from Total Health Conferencing (honorarium), Rocky Mountain Oncology Society (honorarium), and Clinical Care Options (honorarium), other support from Amgen (travel support), Bristol Myers Squibb (travel support), EMD Serono (travel support), Fujifilm (travel support), Millennium (travel support), Sarah Cannon Research Institute (travel support), Synthorx/Sanofi (travel support), GSK (travel support), and Cyteir (travel support), and other support from 3-V Biosciences, Abbisko, AbbVie, ABL Bio, ADC Therapeutics, Accutar, Agenus, Aileron, Alterome, American Society of Clinical Oncology, Amgen, Arcus, ARMO/Eli Lilly and Company, Artios, Astellas, AstraZeneca, Bayer, BeiGene, Beijing Avistone, Bioatla, Bioinvent, Biomea Fusion, Biothera, Bicycle, Black Diamond, Boehringer Ingelheim, Boundless, Celgene, Celldex, Centessa, Ciclomed, Conjupro, Curegenix, Curis, Cyteir, Cytomx, D3 Bio, Daiichi Sankyo, Deciphera, DelMar, Dynamicure, eFFECTOR, Eikon, Eli Lilly and Company, EMD Serono, Epizyme, Erasca, Exelixis, Freenome, Fujifilm, Genmab, GlaxoSmithKline, Harbour BioMed, Hutchison MediPharma, IGM Biosciences, IDEAYA, Ignyta, Ikena, Immuneering, Immunitas, ImmunoGen/MacroGenics, Incyte, Jacobio, Jazz, Jounce, Jubilant, Kineta, Kolltan, Kumquat, Kura, Loxo/Bayer, Medilink, MedImmune, Merck, Metabomed, Millennium, Mirati, miRNA Therapeutics, ModeX, Molecular Templates, Nammi, NIH, Navire/BridgeBio, NGM Bio, NiKang, Novartis, Nuvalent, Nuvectis, OncoMed, Oncusp, Oncorus, Oncothyreon, OnKure, Phanes, Poseida, Precision Oncology, Prelude, PureTech, Pyramid, Pyxis, Quanta, RasCal, Regeneron, Relay, Rgenix, Ribon, Roche, Samumed, Sapience, Sarah Cannon Development Innovations, Seagen, Silicon/Stingthera, Simcha, Sirnaomics, Strategia, Syndax, Synthorx/Sanofi, Taiho, Tachyon, Takeda, Tallac, Tango, Tarus, Tarveda, Teneobio, Tesaro, Tocagen, TORL, Turning Point, University of Texas MD Anderson Cancer Center, Vegenics, Xencor, and Zhuhai Yufan. J.W. Goldman reports grants from Tango Therapeutics during the conduct of the study, as well as grants and personal fees from AstraZeneca, AbbVie, and Eli Lilly and Company, personal fees from Genentech, and grants from Bristol Myers Squibb and Agenus outside the submitted work. A.I. Spira reports grants from Tango Therapeutics during the conduct of the study, as well as grants from Tango Therapeutics outside the submitted work. S.R. Punekar reports nonfinancial support from Tango Therapeutics during the conduct of the study, as well as nonfinancial support from Tango Therapeutics outside the submitted work. D.R. Spigel reports grants from Tango Therapeutics during the conduct of the study, as well as grants and other support from AbbVie, AstraZeneca, Roche/Genentech, GlaxoSmithKline, and Lyell Immunopharma, grants from Agios, Arcus, Ascendis Pharma, Asher Biotherapeutics, BeiGene, Beijing Avistone Biotechnology, Bicara Therapeutics, BioAtla, Blueprint Medicine, Boehringer Ingelheim, Bristol Myers Squibb, Celgene, Chugai, Cyteir Therapeutics, Ellipses Pharma, Erasca, Gilead Sciences, Janux Therapeutics, Jazz Pharmaceuticals, Kronos Bio, Kumquat Biosciences, Loxo Oncology, MacroGenics, Merck, Millennium Pharmaceuticals, Moderna, Molecular Template, Monte Rosa Therapeutics, NGM Biopharmaceuticals, Peloton Therapeutics, Phanes Therapeutics, ProfoundBio, PureTech Health, Razor Genomics, Repare Therapeutics, Rgenix, Scorpion Therapeutics, Shenzhen Chipscreen Biosciences, Stemline Therapeutics, Synthekine, Taiho, Tango Therapeutics, and Zai Laboratory, and other support from Amgen, Circle Pharma, Daiichi Sankyo, Gilead Sciences, MedImmune, ModeX Therapeutics, Ottimo Pharma, and Pyxis Oncology outside the submitted work. J.S. Wang reports other support from Tango Therapeutics during the conduct of the study, as well as other support from AbbVie, Abdera Therapeutics, Accent Therapeutics, Accutar Biotech, Acrivon Therapeutics, Adagene, Allorion Therapeutics, Alterome Therapeutics, Apollo, Artios, Astellas Pharma, BeiGene, Bicycle Therapeutics, BioNTech SE, Biostar, Blueprint Medicines, Bristol Myers Squibb GmbH & Co. KG, Boehringer Ingelheim, C4 Therapeutics, Celgene/Bristol Myers Squibb, Circle Pharma, Compass Therapeutics, Compugen, Cullinan Oncology, Conjupro Biotherapeutics, D3 Bio, Daiichi Sankyo/UCB Japan, Day One Bio, Dren Bio, DualityBio, Edgewood Oncology, Ellipses Pharma, Erasca, Inc, Genentech/Roche, Genmab, Georgiamune, GlaxoSmithKline, Halda Therapeutics, Hotspot Therapeutics, IgM Biosciences, Immunitas, Immunogen, Incyte, ITeos Therapeutics, Janssen, Jazz Pharmaceuticals, Kineta, Klus Pharma, Kumquat, Kura Oncology, Loxo/Lilly, MabSpace Biosciences, Macrogenics, MBQ Pharma, Medikine, MediLink Therapeutics, Stemline/Menarini, Merck KGaA, Mersana, Moderna Therapeutics, NGM Biopharmaceuticals, NiKang, Novartis, Nurix, Olema Oncology, OnCusp Therapeutics, Pfizer, Pyxis, Quanta Therapeutics, Relay Therapeutics, Revolution Medicines, Sanofi, Step Pharma, Syndax, Systimmune, Vividion Therapeutics, Xencor, Zai Lab, and Zymeworks. F. Skoulidis reports grants, personal fees, and other support from Amgen, Revolution Medicines, and Bristol Myers Squibb, grants and personal fees from Merck & Co and Novartis, personal fees from BridgeBio, BeiGene, BergenBio, Guardant Health, Calithera Biosciences, Tango Therapeutics, Roche, Novocure, Hookipa Pharma, Regeneron, ESMO, Japanese Lung Cancer Society, Medscape LLC, Intellisphere LLC, personal fees and other support from AstraZeneca, AACR, IASLC, MJH Life Sciences, IDEOlogy Health, MI&T, PER LLC, and CURIO LLC, and other support from DAVA Oncology outside the submitted work. Y. Yu reports personal fees from Tango Therapeutics during the conduct of the study, as well as ownership of Tango Therapeutics stocks. P. McCarren reports other support from Tango Therapeutics during the conduct of the study. A. Tsai reports personal fees from Tango Therapeutics during the conduct of the study. P. Shahagadkar reports personal fees from Tango Therapeutics during the conduct of the study. N.M. Das reports personal fees from Tango Therapeutics during the conduct of the study. L. Danielczyk reports other support from Tango Therapeutics outside the submitted work. S.R. Meier reports other support from Tango Therapeutics outside the submitted work. D.A. Whittington reports personal fees and other support from Sesame Therapeutics outside the submitted work. C. Min reports other support from Tango Therapeutics outside the submitted work, as well as a patent for WO2024030659A1 pending. I. Sienczylo reports personal fees from Tango Therapeutics during the conduct of the study, as well as a patent 631137 pending. J.P. Maxwell reports personal fees and nonfinancial support from Tango Therapeutics and nonfinancial support from Sesame Therapeutics outside the submitted work, as well as a patent for US 12043607 issued. H.J. DiBenedetto reports other support from Tango Therapeutics during the conduct of the study. A. Crystal reports personal fees and other support from Tango Therapeutics during the conduct of the study, as well as personal fees and other support from Tango Therapeutics outside the submitted work. J.N. Andersen reports personal fees and other support from Tango therapeutics outside the submitted work. K.-K. Wong reports grants from Tango Therapeutics during the conduct of the study, as well as grants from Janssen Pharmaceuticals, Pfizer, Bristol Myers Squibb, Zentalis Pharmaceuticals, Blueprint Pharmaceuticals, Takeda, Novartis, Genentech, BridgeBio Pharma, Boehringer Ingelheim, Cogent, Revolution Medicine, and AstraZeneca and personal fees from Allerion Therapeutics outside the submitted work. No disclosures were reported by the other authors.

Figures

Figure 1.
Figure 1.
HDAC1 knockout resensitizes STK11-mutant tumors to immune checkpoint blockade. A, Average volumes of MC38_sgNTC tumors in the indicated mouse strain; anti–PD-1 antibody was administered biweekly (N = 8 mice/group). B, Average volume of MC38_sgStk11 tumors in the indicated mouse strain; anti–PD-1 antibody was administered biweekly (N = 8 mice/group). Values are mean ± SEM. One-way ANOVA was used to determine statistical significance. **, P < 0.01; ***, P < 0.001; ns, not significant. C, Scheme describing CRISPR screen using STK11 isogenic MC38 tumors. D, Volcano plot of sgRNAs comparing MC38_sgStk11 tumors in C57BL/6 mice treated with anti–PD-1 with MC38_sgStk11 tumors in athymic nude mice. The green point indicates average sgRNAs for HDAC1. E, Volcano plot of sgRNAs comparing MC38_sgStk11 tumors with MC38_sgNTC tumors in C57BL/6 mice treated with anti–PD-1. The green point indicates average sgRNAs for HDAC1. C, Created in BioRender. Ahronian, L. (2025) https://BioRender.com/mndvf20.
Figure 2.
Figure 2.
TNG260 is a CoREST-selective deacetylase inhibitor. A, Structure of TNG260 aligned with structures of class-I HDAC small-molecule inhibitors; the fluorophenyl substituent is highlighted by the dotted lines. B, Crystal structure of TNG260 bound to HDAC2, with the key hydrogen bond and metal interactions illustrated as dotted lines. C, TNG260’s selectivity for HDAC1/2 over HDAC3 can be understood from the overlay of the reported HDAC3 crystal structure (PDB 4A69, blue) onto the TNG260-bound HDAC2 structure (PDB 9NTB, white). HDAC3’s Leu133 position clashes with the TNG260 fluorophenyl group, whereas HDAC2’s Leu140 position allows tight ligand binding but would clash with HDAC3’s Tyr107. D, Inhibition of the indicated HDAC enzyme in Fluor de Lys deacetylase assay by 3 hours of incubation with a dose response of TNG260. Plots are averages of N = 2 experiments. E, Western blotting of acetyl-H3K9 in the MC38 cell line treated with a dose response of TNG260 for 72 hours. F, Cellular HDAC1, HDAC2, and HDAC3 NanoBRET assay with a dose response of TNG260 incubated for 3 hours. Plots are averages of N = 2 experiments. G, Heat map showing IC50s of purified HDAC complexes profiled in an in vitro deacetylase assay with the indicated compounds.
Figure 3.
Figure 3.
TNG260 reverses resistance to anti–PD-1 in STK11-deficient tumor models. A, Mouse plasma levels of unbound (u) TNG260 after 2 days of once daily oral treatment with TNG260 at the indicated dose (N = 4 mice/group). The IC50 of HDAC1 in the cellular NanoBRET assay is indicated by the dotted red line. B, Relative amounts of acetyl-H3K9 normalized to histone 3 from tumor tissue at the doses indicated, quantified from Western blotting. Each time point is normalized to the vehicle control tumors (N = 4 mice/group). C, Relative levels of acetyl-H3K9 normalized to histone H3 following 7 days of TNG260 treatment at the indicated dose, quantified from Western blotting (N = 3 representative tumors/group). Values are average ± SD. A t test was used for statistical analysis comparing each treatment with vehicle control. D, Individual MC38_sgStk11 tumor volumes treated with anti-IgG2a and TNG260 (once daily orally) at 30 mg/kg, anti–PD-1 (intraperitoneally twice weekly) at 10 mg/kg, or the combination of TNG260 and anti–PD-1 (N = 8 mice/group). ORR was calculated from the sum of mice with a partial regression and complete regression. Statistical analysis was performed by one-way ANOVA for the combination arm versus anti–PD-1. E and F, Survival curve of the indicated treatment groups in the MC38_sgStk11 study from D (E) or CT26_sgStk11 from Supplementary Fig. S4 (F). Statistical analysis was performed comparing the combination TNG260 and anti–PD-1 groups versus the anti–PD-1 groups by log-rank tests. G, Inhibition of HDAC1 and HDAC3 as determined by cellular NanoBRET assay plotted by free exposures of TNG260 in vivo. Shaded box indicates efficacious exposures of TNG260 in mouse. H, Average tumor volumes of animals previously cured of MC38_sgStk11 tumors with combination treatment (N = 5) compared with an untreated, naïve cohort (N = 8). Mice were rechallenged with MC38_sgStk11 tumor cells and tumor volume was evaluated over time. I and J, Average tumor volumes of MC38_sgStk11 tumors in C57BL/6 (I) and BALB/c athymic nude mice (J) treated as indicated (N = 8 mice/group). Statistical comparisons were carried out by one-way ANOVA comparing anti-IgG2a to each treatment group. *, P ≤ 0.05; **, P ≤ 0.01; ***, P ≤ 0.001; ****, P ≤ 0.0001; ns, not significant.
Figure 4.
Figure 4.
TNG260 sensitizes STK11-deficient lung tumors to anti–PD-1 treatment through transcriptional reprogramming. A and B, Average tumor volumes of KL tumors (N = 15–20 mice/group; A) and KP tumors (N = 5 mice/group; B) in B6-Albino mice, treated as indicated. C, Tumor volume changes measured by MRI scans of cre-induced KL GEMM mice, treated as indicated (N = 8–10 mice/group). Statistical comparisons were carried out by one-way ANOVA comparing anti-IgG2a with each treatment group. **, P ≤ 0.01; ***, P ≤ 0.001; ****, P ≤ 0.0001; ns, not significant. D, Bar chart showing fold change of the expression of genes related to antigen presentation and peptide loading in KL and KP cells treated with 1 µmol/L TNG260 for 7 days. E, Heatmap showing acetyl-H3K9 signal in both KL and KP cell lines treated with DMSO (control) or 1 µmol/L TNG260 (N = 2) for 7 days. ChIP signal intensity is shown by red shading. Genomic regions are clustered into seven groups by k-means (k = 7) clustering. F, Genome browser view at the Cxcl3 locus of KL and KP cell lines treated with DMSO or TNG260 (N = 2). G, Enrichment plot showing positive enrichment of differentially accessible acetyl-H3K9 peaks for genes increased by TNG260 treatment. H, GO analysis of genes from KL tumors in clusters 3 and 4 from Supplementary Fig. S7I. Clusters 3 and 4 were defined as genes with increased acetyl-H3K9 and increased expression with TNG260 treatment. I, Heatmap showing acetyl-H3K9 signals in KL and KP tumors treated with the combination of TNG260 and anti–PD-1. ChIP signal intensity is shown by red shading. Genomic regions are clustered into seven groups by k-means (k = 6) clustering (N = 3–5 mice/group).
Figure 5.
Figure 5.
TNG260 modifies the tumor immune microenvironment in patients with STK11-deficient tumors. A, Percentage of acetylated histone H4 Lys5 compared with total acetylated and unacetylated histone H4 detected by mass spectrometry in FFPE tissue collected before and in study. B, TPS of PD-L1 from FFPE tissue biopsies collected in A. C and D, FFPE tissue biopsies analyzed by multiplex immunofluorescence for helper T cells (CD3+/CD4+; C) and cytotoxic T cells (CD3+/CD8+; D), respectively. Quantifications were performed in tumor regions identified by hematoxylin and eosin. E, Exemplar paired biopsy of a patient with NSCLC (patient 9; Supplementary Table S6) imaged by multiplex immunofluorescence for the indicated markers before and during treatment with 120 mg TNG260 and pembrolizumab. Scale bar, 50 µm. *, P ≤ 0.05.

References

    1. Ricciuti B, Arbour KC, Lin JJ, Vajdi A, Vokes N, Hong L, et al. Diminished efficacy of programmed death-(ligand)1 inhibition in STK11- and KEAP1-mutant lung adenocarcinoma is affected by KRAS mutation status. J Thorac Oncol 2022;17:399–410. - PMC - PubMed
    1. Alexander B, Sokol ES, Danziger NA, Pavlick DC, Elvin J, Killian JK, et al. 107P immune checkpoint inhibitor (ICPI) resistance genes STK11 and KEAP1: a comparative comprehensive genomic profiling (CGP) study. Ann Oncol 2020;31:S283–4.
    1. Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 2013;6:pl1. - PMC - PubMed
    1. Cooper AJ, Muzikansky A, Lennerz J, Narinesingh F, Mino-Kenudson M, Hung YP, et al. Clinicopathologic characteristics and outcomes for patients with KRAS G12D-mutant NSCLC. JTO Clin Res Rep 2022;3:100390. - PMC - PubMed
    1. Heist RS, Yu J, Donderici EY, Zhang N, Espenschied CR, Lang K, et al. Impact of STK11 mutation on first-line immune checkpoint inhibitor (ICI) outcomes in a real-world KRAS G12C mutant lung adenocarcinoma cohort. J Clin Oncol 2021;39:9106.

MeSH terms

Substances