Distinct molecular subtypes of KRASG12C-mutant lung adenocarcinoma: Insights into clinical outcomes, tumour microenvironments and therapeutic strategies

Haitang Yang , Anshun Zhu , Yongliang Niu , Wenyan Ma , Ke Xu , Yunxuan Jia , Weijiao Xu , Baicheng Zhao , Enshuo Zhang , Jiaying Jia , Shunqing Liang , Patrick Dorn , Gang Liu , Ren-Wang Peng , Feng Yao

Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (10) : e70490

PDF
Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (10) : e70490 DOI: 10.1002/ctm2.70490
RESEARCH ARTICLE

Distinct molecular subtypes of KRASG12C-mutant lung adenocarcinoma: Insights into clinical outcomes, tumour microenvironments and therapeutic strategies

Author information +
History +
PDF

Abstract

Background: KRASG12C is the most common KRAS mutation in lung adenocarcinoma (LUAD), yet clinical responses to KRASG12C-selective inhibitors (G12Ci) and immunotherapy remain variable.

Methods: Transcriptomic analysis of KRASG12C-mutant LUAD was performed using machine learning algorithms to classify molecular subtypes. Subtype-specific features, including genomic alterations, tumour microenvironment and therapeutic vulnerabilities, were systematically evaluated.

Results: We identified three distinct molecular subtypes (KC1, KC2 and KC3) of KRASG12C-mutant LUAD through transcriptomic analysis using machine learning algorithms. KC1 subtype is characterised by a neuroendocrine phenotype associated with SMARCA4 loss-of-function and frequent STK11 co-mutations, with a relatively good prognosis. It exhibits poor immune infiltration and demonstrates resistance to G12Ci and immunotherapy but shows sensitivity to MEK1/2 inhibitors; KC2 subtype exhibits a highly malignant phenotype with high proliferation, increased glucose metabolism, and the poorest prognosis. It is enriched with T-cell infiltration and responds best to G12Ci monotherapy and immunotherapy. KC3 subtype is distinguished by well differentiation and the best survival, with an immune-enriched microenvironment featuring abundant immune-suppressive cancer-associated fibroblasts. It demonstrates limited sensitivity to G12Ci and a moderate response to immunotherapy. Notably, KC1‒3 subtype-specific molecular signatures predict drug sensitivity more accurately than classical KRASG12C signalling models.

Conclusions: These findings illuminate the intricate interplay between tumour subtypes, microenvironmental factors and therapeutic responses, offering a robust framework for improved patient stratification and the development of personalised therapeutic strategies KRASG12C-mutant LUAD.

Keywords

KRAS mutation / lung adenocarcinoma / molecular subtypes / precision oncology / treatment response / tumour immune microenvironment

Cite this article

Download citation ▾
Haitang Yang, Anshun Zhu, Yongliang Niu, Wenyan Ma, Ke Xu, Yunxuan Jia, Weijiao Xu, Baicheng Zhao, Enshuo Zhang, Jiaying Jia, Shunqing Liang, Patrick Dorn, Gang Liu, Ren-Wang Peng, Feng Yao. Distinct molecular subtypes of KRASG12C-mutant lung adenocarcinoma: Insights into clinical outcomes, tumour microenvironments and therapeutic strategies. Clinical and Translational Medicine, 2025, 15(10): e70490 DOI:10.1002/ctm2.70490

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Arbour KC, Ricciuti B, Rizvi H, et al. Chemo-immunotherapy outcomes of KRAS-G12C mutant lung cancer compared to other molecular subtypes of KRAS-mutant lung cancer. J Clin Oncol. 2021; 39(suppl 15): 9088.

[2]

Gadgeel S, Rodriguez-Abreu D, Felip E, et al. KRAS mutational status and efficacy in KEYNOTE-189: pembrolizumab (pembro) plus chemotherapy (chemo) vs placebo plus chemo as first-line therapy for metastatic non-squamous NSCLC. Ann Oncol. 2019; 30: xi64-xi65.

[3]

Yang H, Liang SQ, Schmid RA, Peng RW. New horizons in KRAS-mutant lung cancer: dawn after darkness. Front Oncol. 2019; 9: 953.

[4]

Fakih MG, Kopetz S, Kuboki Y, et al. Sotorasib for previously treated colorectal cancers with KRAS(G12C) mutation (CodeBreaK100): a prespecified analysis of a single-arm, phase 2 trial. Lancet Oncol. 2022; 23(1): 115-124.

[5]

Hong DS, Fakih MG, Strickler JH, et al. KRAS(G12C) inhibition with sotorasib in advanced solid tumors. New Engl J Med. 2020; 383(13): 1207-1217.

[6]

Jänne PA, Riely GJ, Gadgeel SM, et al. Adagrasib in non-small-cell lung cancer harboring a KRAS(G12C) mutation. New Engl J Med. 2022; 387(2): 120-131.

[7]

Skoulidis F, Li BT, Dy GK, et al. Sotorasib for lung cancers with KRAS p.G12C mutation. New Engl J Med. 2021; 384(25): 2371-2381.

[8]

Xue JY, Zhao Y, Aronowitz J, et al. Rapid non-uniform adaptation to conformation-specific KRAS(G12C) inhibition. Nature. 2020; 577(7790): 421-425.

[9]

Awad MM, Liu S, Rybkin II, et al. Acquired resistance to KRAS(G12C) inhibition in cancer. New Engl J Med. 2021; 384(25): 2382-2393.

[10]

Tsai YS, Woodcock MG, Azam SH, et al. Rapid idiosyncratic mechanisms of clinical resistance to KRAS G12C inhibition. J Clin Invest. 2022; 132(4): e155523.

[11]

Akhave NS, Biter AB, Hong DS. Mechanisms of resistance to KRAS(G12C)-targeted therapy. Cancer Discov. 2021; 11(6): 1345-1352.

[12]

Multiple mechanisms underlie the acquired resistance to KRAS G12C inhibition. Cancer Discov. 2022; 12(3): OF7.

[13]

Wang XD, Lin JH, Hu MH. Discovery of a tribenzophenazine analog for binding to the KRAS mRNA G-quadruplex structures in the cisplatin-resistant non-small cell lung cancer. J Biol Chem. 2025; 301(2): 108164.

[14]

Skoulidis F, Li BT, de Langen AJ, et al. Molecular determinants of sotorasib clinical efficacy in KRAS(G12C)-mutated non-small-cell lung cancer. Nat Med. 2025; 31(8): 2755-2767.

[15]

Mugarza E, van Maldegem F, Boumelha J, et al. Therapeutic KRAS(G12C) inhibition drives effective interferon-mediated antitumor immunity in immunogenic lung cancers. Sci Adv. 2022; 8(29): eabm8780.

[16]

Canon J, Rex K, Saiki AY, et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature. 2019; 575(7781): 217-223.

[17]

Wilkerson MD, Yin X, Walter V, et al. Differential pathogenesis of lung adenocarcinoma subtypes involving sequence mutations, copy number, chromosomal instability, and methylation. PloS ONE. 2012; 7(5): e36530.

[18]

Skoulidis F, Byers LA, Diao L, et al. Co-occurring genomic alterations define major subsets of KRAS-mutant lung adenocarcinoma with distinct biology, immune profiles, and therapeutic vulnerabilities. Cancer Discov. 2015; 5(8): 860-877.

[19]

He L, Kulesskiy E, Saarela J, et al. Methods for high-throughput drug combination screening and synergy scoring. Methods Mol Biol. 2018; 1711: 351-398.

[20]

Yang H, Sun B, Ma W, et al. Multi-scale characterization of tumor-draining lymph nodes in resectable lung cancer treated with neoadjuvant immune checkpoint inhibitors. EBioMedicine. 2022; 84: 104265.

[21]

Yang H, Sun B, Xu K, et al. Pharmaco-transcriptomic correlation analysis reveals novel responsive signatures to HDAC inhibitors and identifies Dasatinib as a synergistic interactor in small-cell lung cancer. EBioMedicine. 2021; 69: 103457.

[22]

Wang L, Yang H, Dorn P, et al. Peritumoral CD90+CD73+ cells possess immunosuppressive features in human non-small cell lung cancer. EBioMedicine. 2021; 73: 103664.

[23]

Lee DD, Seung HS. Learning the parts of objects by non-negative matrix factorization. Nature. 1999; 401(6755): 788-791.

[24]

Gao Y, Church G. Improving molecular cancer class discovery through sparse non-negative matrix factorization. Bioinformatics. 2005; 21(21): 3970-3975.

[25]

Tong X, Patel AS, Kim E, et al. Adeno-to-squamous transition drives resistance to KRAS inhibition in LKB1 mutant lung cancer. Cancer Cell. 2024; 42(3): 413-428.e7.

[26]

Rudin CM, Poirier JT, Byers LA, et al. Molecular subtypes of small cell lung cancer: a synthesis of human and mouse model data. Nat Rev Cancer. 2019; 19(5): 289-297.

[27]

Ireland AS, Micinski AM, Kastner DW, et al. MYC drives temporal evolution of small cell lung cancer subtypes by reprogramming neuroendocrine fate. Cancer Cell. 2020; 38(1): 60-78.e12.

[28]

Voigt E, Wallenburg M, Wollenzien H, et al. Sox2 is an oncogenic driver of small-cell lung cancer and promotes the classic neuroendocrine subtype. Mol Cancer Res. 2021; 19(12): 2015-2025.

[29]

Augustyn A, Borromeo M, Wang T, et al. ASCL1 is a lineage oncogene providing therapeutic targets for high-grade neuroendocrine lung cancers. Proc Natl Acad Sci U S A. 2014; 111(41): 14788-14793.

[30]

Kosari F, Ida CM, Aubry MC, et al. ASCL1 and RET expression defines a clinically relevant subgroup of lung adenocarcinoma characterized by neuroendocrine differentiation. Oncogene. 2014; 33(29): 3776-3783.

[31]

Miyashita N, Horie M, Suzuki HI, et al. An integrative analysis of transcriptome and epigenome features of ASCL1-positive lung adenocarcinomas. J Thorac Oncol. 2018; 13(11): 1676-1691.

[32]

Carnaghi C, Rimassa L, Garassino I, Santoro A. Clinical significance of neuroendocrine phenotype in non-small-cell lung cancer. Ann Oncol. 2001(12 suppl 2): S119-S123.

[33]

Ionescu DN, Treaba D, Gilks CB, et al. Nonsmall cell lung carcinoma with neuroendocrine differentiation–an entity of no clinical or prognostic significance. Am J Surg Pathol. 2007; 31(1): 26-32.

[34]

Cheung WK, Zhao M, Liu Z, et al. Control of alveolar differentiation by the lineage transcription factors GATA6 and HOPX inhibits lung adenocarcinoma metastasis. Cancer Cell. 2013; 23(6): 725-738.

[35]

Yang B, Zhang W, Zhang M, et al. KRT6A promotes EMT and cancer stem cell transformation in lung adenocarcinoma. Technol Cancer Res Treat. 2020; 19: 1533033820921248.

[36]

He Y, Luo W, Liu Y, et al. IL-20RB mediates tumoral response to osteoclastic niches and promotes bone metastasis of lung cancer. J Clin Invest. 2022; 132(20): e157917.

[37]

Jain R, Barkauskas CE, Takeda N, et al. Plasticity of Hopx(+) type I alveolar cells to regenerate type II cells in the lung. Nat Commun. 2015; 6: 6727.

[38]

Yang H. Co-occurring LKB1 deficiency determinates the susceptibility to ERK-targeted therapy in RAS-mutant lung cancer. J Thorac Oncol. 2020; 15(4): e58-59.

[39]

Arbour KC, Jordan E, Kim HR, et al. Effects of co-occurring genomic alterations on outcomes in patients with KRAS-mutant non-small cell lung cancer. Clin Cancer Res. 2018; 24(2): 334-340.

[40]

Skoulidis F, Goldberg ME, Greenawalt DM, et al. STK11/LKB1 mutations and PD-1 inhibitor resistance in KRAS-mutant lung adenocarcinoma. Cancer Discov. 2018; 8(7): 822-835.

[41]

Negrao MV, Araujo HA, Lamberti G, et al. Comutations and KRASG12C inhibitor efficacy in advanced NSCLC. Cancer Discov. 2023; 13(7): 1556-1571.

[42]

Arbour KC, Rizvi H, Plodkowski AJ, et al. Treatment outcomes and clinical characteristics of patients with KRAS-G12C-mutant non-small cell lung cancer. Clin Cancer Res. 2021; 27(8): 2209-2215.

[43]

Salem ME, El-Refai SM, Sha W, et al. Landscape of KRAS(G12C), associated genomic alterations, and interrelation with immuno-oncology biomarkers in KRAS-mutated cancers. JCO Precis Oncol. 2022; 6: e2100245.

[44]

Pirlog R, Piton N, Lamy A, et al. Morphological and molecular characterization of KRAS G12C-mutated lung adenocarcinomas. Cancers. 2022; 14(4): 1030.

[45]

Kumarasamy V, Wang J, Frangou C, et al. The extracellular niche and tumor microenvironment enhance KRAS inhibitor efficacy in pancreatic cancer. Cancer Res. 2024; 84(7): 1115-1132.

[46]

Hu H, Cheng R, Wang Y, et al. Oncogenic KRAS signaling drives evasion of innate immune surveillance in lung adenocarcinoma by activating CD47. J Clin Invest. 2023; 133(2): e153470.

[47]

Slyper M, Porter CBM, Ashenberg O, et al. A single-cell and single-nucleus RNA-seq toolbox for fresh and frozen human tumors. Nat Med. 2020; 26(5): 792-802.

[48]

Pabla S, Conroy JM, Nesline MK, et al. Proliferative potential and resistance to immune checkpoint blockade in lung cancer patients. J Immunother Cancer. 2019; 7(1): 27.

[49]

Thorsson V, Gibbs DL, Brown SD, et al. The immune landscape of cancer. Immunity. 2018; 48(4): 812-830.e14.

[50]

Bill R, Wirapati P, Messemaker M, et al. CXCL9:sPP1 macrophage polarity identifies a network of cellular programs that control human cancers. Science. 2023; 381(6657): 515-524.

[51]

Bagaev A, Kotlov N, Nomie K, et al. Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. Cancer Cell. 2021; 39(6): 845-865.e7.

[52]

Altorki NK, Bhinder B, Borczuk AC, et al. A signature of enhanced proliferation associated with response and survival to anti-PD-L1 therapy in early-stage non-small cell lung cancer. Cell Rep Med. 2024; 5(3): 101438.

[53]

Trapnell C, Cacchiarelli D, Grimsby J, et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol. 2014; 32(4): 381-386.

[54]

Arpinati L, Scherz-Shouval R. From gatekeepers to providers: regulation of immune functions by cancer-associated fibroblasts. Trends Cancer. 2023; 9(5): 421-443.

[55]

Templin J, Atanackovic D, Hasche D, Radhakrishnan SV, Luetkens T. Oscillating expression of interleukin-16 in multiple myeloma is associated with proliferation, clonogenic growth, and PI3K/NFKB/MAPK activation. Oncotarget. 2017; 8(30): 49253-49263.

[56]

Dy GK, Govindan R, Velcheti V, et al. Long-term outcomes and molecular correlates of sotorasib efficacy in patients with pretreated KRAS G12C-mutated non-small-cell lung cancer: 2-year analysis of CodeBreaK 100. J Clin Oncol. 2023; 41(18): 3311-3317.

[57]

Jung H, Kim HS, Kim JY, et al. DNA methylation loss promotes immune evasion of tumours with high mutation and copy number load. Nat Commun. 2019; 10(1): 4278.

[58]

Gupta A, Towers C, Willenbrock F, et al. Dual-specificity protein phosphatase DUSP4 regulates response to MEK inhibition in BRAF wild-type melanoma. Br J Cancer. 2020; 122(4): 506-516.

[59]

Ito T, Young MJ, Li R, et al. Paralog knockout profiling identifies DUSP4 and DUSP6 as a digenic dependence in MAPK pathway-driven cancers. Nat Genet. 2021; 53(12): 1664-1672.

[60]

Klomp JA, Klomp JE, Stalnecker CA, et al. Defining the KRAS- and ERK-dependent transcriptome in KRAS-mutant cancers. Science. 2024; 384(6700): eadk0775.

[61]

Klomp JE, Diehl JN, Klomp JA, et al. Determining the ERK-regulated phosphoproteome driving KRAS-mutant cancer. Science. 2024; 384(6700): eadk0850.

[62]

Ryan MB, de la Cruz FF, Phat S, et al. Vertical pathway inhibition overcomes adaptive feedback resistance to KRAS(G12C) inhibition. Clin Cancer Res. 2020; 26(7): 1633-1643.

[63]

Li S, Liu S, Deng J, et al. Assessing therapeutic efficacy of MEK inhibition in a KRAS(G12C)-driven mouse model of lung cancer. Clin Cancer Res. 2018; 24(19): 4854-4864.

[64]

Fujiwara T, Hiramatsu M, Isagawa T, et al. ASCL1-coexpression profiling but not single gene expression profiling defines lung adenocarcinomas of neuroendocrine nature with poor prognosis. Lung Cancer. 2012; 75(1): 119-125.

[65]

Rekhtman N, Montecalvo J, Chang JC, et al. SMARCA4-deficient thoracic sarcomatoid tumors represent primarily smoking-related undifferentiated carcinomas rather than primary thoracic sarcomas. J Thorac Oncol. 2020; 15(2): 231-247.

[66]

Agaimy A, Fuchs F, Moskalev EA, et al. SMARCA4-deficient pulmonary adenocarcinoma: clinicopathological, immunohistochemical, and molecular characteristics of a novel aggressive neoplasm with a consistent TTF1(neg)/CK7(pos)/HepPar-1(pos) immunophenotype. Virchows Arch. 2017; 471(5): 599-609.

[67]

Hiroshima K, Iyoda A, Shibuya K, et al. Prognostic significance of neuroendocrine differentiation in adenocarcinoma of the lung. Ann Thorac Surg. 2002; 73(6): 1732-1735.

[68]

Kriegsmann K, Zgorzelski C, Muley T, et al. Role of synaptophysin, chromogranin and CD56 in adenocarcinoma and squamous cell carcinoma of the lung lacking morphological features of neuroendocrine differentiation: a retrospective large-scale study on 1170 tissue samples. BMC Cancer. 2021; 21(1): 486.

[69]

Schoenfeld AJ, Bandlamudi C, Lavery JA, et al. The genomic landscape of SMARCA4 alterations and associations with outcomes in patients with lung cancer. Clin Cancer Res. 2020; 26(21): 5701-5708.

[70]

Wang Y, Meraz IM, Qudratullah M, et al. Mutation of SMARCA4 induces cancer cell-intrinsic defects in the enhancer landscape and resistance to immunotherapy. Cancer Res. 2025; 85(11): 1997-2013.

[71]

Davar D, Wang H, Chauvin JM, et al. Phase Ib/II study of pembrolizumab and pegylated-interferon alfa-2b in advanced melanoma. J Clin Oncol. 2018; 36(35): Jco1800632.

[72]

Fernández-García F, Fernández-Rodríguez A, Fustero-Torre C, et al. Type I interferon signaling pathway enhances immune-checkpoint inhibition in KRAS mutant lung tumors. Proc Natl Acad Sci U S A. 2024; 121(36): e2402913121.

RIGHTS & PERMISSIONS

2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

17

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/