Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Scribble mis-localization induces adaptive resistance to KRAS G12C inhibitors through feedback activation of MAPK signaling mediated by YAP-induced MRAS

Abstract

Tumor cells evade targeted drugs by rewiring their genetic and epigenetic networks. Here, we identified that inhibition of MAPK signaling rapidly induces an epithelial-to-mesenchymal transition program by promoting re-localization of an apical-basal polarity protein, Scribble, in oncogene-addicted lung cancer models. Mis-localization of Scribble suppressed Hippo-YAP signaling, leading to YAP nuclear translocation. Furthermore, we discovered that a RAS superfamily protein MRAS is a direct target of YAP. Treatment with KRAS G12C inhibitors induced MRAS expression, which formed a complex with SHOC2, precipitating feedback activation of MAPK signaling. Abrogation of YAP activation or MRAS induction enhanced the efficacy of KRAS G12C inhibitor treatment in vivo. These results highlight a role for protein localization in the induction of a non-genetic mechanism of resistance to targeted therapies in lung cancer. Furthermore, we demonstrate that induced MRAS expression is a key mechanism of adaptive resistance following KRAS G12C inhibitor treatment.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Scrib mis-localization following KRAS G12C inhibition is a cause of resistance to KRAS G12C inhibitors.
Fig. 2: Inhibition of the MAPK pathway induces Scrib mis-localization.
Fig. 3: ZDHHC7 is involved in the regulation of Scrib localization.
Fig. 4: Scrib mis-localization induces nuclear translocation of YAP, leading to adaptive resistance to KRAS G12C inhibitors.
Fig. 5: MRAS is transcriptionally regulated by YAP.
Fig. 6: KRAS G12C inhibition rewires signaling from a Scrib–SHOC2 complex to an MRAS–SHOC2 complex, leading to reactivation of MAPK signaling.
Fig. 7: Membrane-localized MRAS is activated by receptor tyrosine kinases.
Fig. 8: MRAS knockout enhances sotorasib efficacy in vivo.

Similar content being viewed by others

Data availability

RNA-sequencing data generated in this study have been deposited in the BioProject database at DNA Data Bank of Japan under accession code DRA014731. Source data for Figs. 3e,f, 4c,h, 5b,f–h, 6e, 7a,b and 8a and Extended Data Figs. 1e,f,h–j, 3b,e,k,l, 6a,c,d, 8a–c and 9a–c,g have been provided as Source Data files. All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

References

  1. Prior, I. A., Hood, F. E. & Hartley, J. L. The frequency of ras mutations in cancer. Cancer Res. 80, 2969–2974 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Ostrem, J. M., Peters, U., Sos, M. L., Wells, J. A. & Shokat, K. M. K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature 503, 548–551 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Canon, J. et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature 575, 217–223 (2019).

    Article  CAS  PubMed  Google Scholar 

  4. Hallin, J. et al. The KRAS(G12C) inhibitor MRTX849 provides insight toward therapeutic susceptibility of KRAS-mutant cancers in mouse models and patients. Cancer Discov. 10, 54–71 (2020).

    Article  CAS  PubMed  Google Scholar 

  5. Janes, M. R. et al. Targeting KRAS mutant cancers with a covalent G12C-specific inhibitor. Cell 172, 578–589 (2018).

    Article  CAS  PubMed  Google Scholar 

  6. Skoulidis, F. et al. Sotorasib for lung cancers with KRAS p.G12C mutation. N. Engl. J. Med. 384, 2371–2381 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Janne, P. A. et al. Adagrasib in non-small-cell lung cancer harboring a KRAS(G12C) mutation. N. Engl. J. Med. https://doi.org/10.1056/NEJMoa2204619 (2022).

  8. de Langen, A. J. et al. Sotorasib versus docetaxel for previously treated non-small-cell lung cancer with KRAS(G12C) mutation: a randomised, open-label, phase 3 trial. Lancet 401, 733–746 (2023).

    Article  PubMed  Google Scholar 

  9. Awad, M. M. et al. Acquired resistance to KRAS(G12C) inhibition in cancer. N. Engl. J. Med. 384, 2382–2393 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zhao, Y. et al. Diverse alterations associated with resistance to KRAS(G12C) inhibition. Nature 599, 679–683 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Adachi, Y. et al. Epithelial-to-mesenchymal transition is a cause of both intrinsic and acquired resistance to KRAS G12C inhibitor in KRAS G12C-mutant non-small cell lung cancer. Clin. Cancer Res. 26, 5962–5973 (2020).

    Article  CAS  PubMed  Google Scholar 

  12. Solanki, H. S. et al. Cell type-specific adaptive signaling responses to KRAS(G12C) inhibition. Clin. Cancer Res. 27, 2533–2548 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Singh, A. et al. A gene expression signature associated with ‘K-Ras addiction’ reveals regulators of EMT and tumor cell survival. Cancer Cell 15, 489–500 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lito, P., Rosen, N. & Solit, D. B. Tumor adaptation and resistance to RAF inhibitors. Nat. Med. 19, 1401–1409 (2013).

    Article  CAS  PubMed  Google Scholar 

  15. Hata, A. N. et al. Tumor cells can follow distinct evolutionary paths to become resistant to epidermal growth factor receptor inhibition. Nat. Med. 22, 262–269 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lito, P., Solomon, M., Li, L. S., Hansen, R. & Rosen, N. Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism. Science 351, 604–608 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kitai, H. et al. Epithelial-to-mesenchymal transition defines feedback activation of receptor tyrosine kinase signaling induced by MEK inhibition in KRAS-mutant lung cancer. Cancer Discov. 6, 754–769 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Manchado, E. et al. A combinatorial strategy for treating KRAS-mutant lung cancer. Nature 534, 647–651 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Misale, S. et al. KRAS G12C NSCLC models are sensitive to direct targeting of KRAS in combination with PI3K inhibition. Clin. Cancer Res. 25, 796–807 (2019).

    Article  CAS  PubMed  Google Scholar 

  20. Ryan, M. B. et al. Vertical pathway inhibition overcomes adaptive feedback resistance to KRAS(G12C) inhibition. Clin. Cancer Res. 26, 1633–1643 (2020).

    Article  CAS  PubMed  Google Scholar 

  21. Aiello, N. M. et al. EMT subtype influences epithelial plasticity and mode of cell migration. Dev. Cell 45, 681–695 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Huang, R. Y., Guilford, P. & Thiery, J. P. Early events in cell adhesion and polarity during epithelial–mesenchymal transition. J. Cell Sci. 125, 4417–4422 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Yang, J. et al. Guidelines and definitions for research on epithelial–mesenchymal transition. Nat. Rev. Mol. Cell Biol. 21, 341–352 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Lee, M. & Vasioukhin, V. Cell polarity and cancer: cell and tissue polarity as a non-canonical tumor suppressor. J. Cell Sci. 121, 1141–1150 (2008).

    Article  CAS  PubMed  Google Scholar 

  25. Hernandez, J. L. et al. APT2 inhibition restores scribble localization and S-palmitoylation in snail-transformed cells. Cell Chem. Biol. 24, 87–97 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Chen, B. et al. ZDHHC7-mediated S-palmitoylation of Scribble regulates cell polarity. Nat. Chem. Biol. 12, 686–693 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ko, P. J. & Dixon, S. J. Protein palmitoylation and cancer. EMBO Rep. https://doi.org/10.15252/embr.201846666 (2018).

  28. Abrami, L. et al. Palmitoylated acyl protein thioesterase APT2 deforms membranes to extract substrate acyl chains. Nat. Chem. Biol. 17, 438–447 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Cordenonsi, M. et al. The Hippo transducer TAZ confers cancer stem cell-related traits on breast cancer cells. Cell 147, 759–772 (2011).

    Article  CAS  PubMed  Google Scholar 

  30. Kapoor, A. et al. Yap1 activation enables bypass of oncogenic Kras addiction in pancreatic cancer. Cell 158, 185–197 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Shao, D. D. et al. KRAS and YAP1 converge to regulate EMT and tumor survival. Cell 158, 171–184 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kaneda, A. et al. The novel potent TEAD inhibitor, K-975, inhibits YAP1/TAZ-TEAD protein-protein interactions and exerts an anti-tumor effect on malignant pleural mesothelioma. Am. J. Cancer Res. 10, 4399–4415 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Moroishi, T., Hansen, C. G. & Guan, K. L. The emerging roles of YAP and TAZ in cancer. Nat. Rev. Cancer 15, 73–79 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Simanshu, D. K., Nissley, D. V. & McCormick, F. RAS proteins and their regulators in human disease. Cell 170, 17–33 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Young, L. C. & Rodriguez-Viciana, P. MRAS: a close but understudied member of the RAS family. Cold Spring Harb. Perspect. Med. https://doi.org/10.1101/cshperspect.a033621 (2018).

  36. Kurppa, K. J. et al. Treatment-induced tumor dormancy through YAP-mediated transcriptional reprogramming of the apoptotic pathway. Cancer Cell 37, 104–122 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Young, L. C. et al. An MRAS, SHOC2, and Scrib complex coordinates ERK pathway activation with polarity and tumorigenic growth. Mol. Cell 52, 679–692 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Young, L. C. et al. SHOC2-MRAS-PP1 complex positively regulates RAF activity and contributes to Noonan syndrome pathogenesis. Proc. Natl Acad. Sci. USA 115, E10576–E10585 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Marine, J. C., Dawson, S. J. & Dawson, M. A. Non-genetic mechanisms of therapeutic resistance in cancer. Nat. Rev. Cancer 20, 743–756 (2020).

    Article  CAS  PubMed  Google Scholar 

  40. Lai, L. P. et al. Classical RAS proteins are not essential for paradoxical ERK activation induced by RAF inhibitors. Proc. Natl Acad. Sci. USA https://doi.org/10.1073/pnas.2113491119 (2022).

  41. Hauseman, Z. J. et al. Structure of the MRAS-SHOC2-PP1C phosphatase complex. Nature 609, 416–423 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kwon, J. J. et al. Structure-function analysis of the SHOC2-MRAS-PP1C holophosphatase complex. Nature 609, 408–415 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Liau, N. P. D. et al. Structural basis for SHOC2 modulation of RAS signalling. Nature 609, 400–407 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Bonsor, D. A. et al. Structure of the SHOC2-MRAS-PP1C complex provides insights into RAF activation and Noonan syndrome. Nat. Struct. Mol. Biol. 29, 966–977 (2022).

    Article  CAS  PubMed  Google Scholar 

  45. Jones, G. G. et al. SHOC2 phosphatase-dependent RAF dimerization mediates resistance to MEK inhibition in RAS-mutant cancers. Nat. Commun. 10, 2532 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Sulahian, R. et al. Synthetic lethal interaction of SHOC2 depletion with MEK inhibition in RAS-driven cancers. Cell Rep. 29, 118–134 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Terai, H. et al. SHOC2 Is a critical modulator of sensitivity to EGFR-TKIs in non-small cell lung cancer cells. Mol. Cancer Res. 19, 317–328 (2021).

    Article  CAS  PubMed  Google Scholar 

  48. Kota, P. et al. M-Ras/Shoc2 signaling modulates E-cadherin turnover and cell–cell adhesion during collective cell migration. Proc. Natl Acad. Sci. USA 116, 3536–3545 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Mathieu, M. E. et al. MRAS GTPase is a novel stemness marker that impacts mouse embryonic stem cell plasticity and Xenopus embryonic cell fate. Development 140, 3311–3322 (2013).

    Article  CAS  PubMed  Google Scholar 

  50. Singh, A. & Settleman, J. EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene 29, 4741–4751 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Dongre, A. & Weinberg, R. A. New insights into the mechanisms of epithelial–mesenchymal transition and implications for cancer. Nat. Rev. Mol. Cell Biol. 20, 69–84 (2019).

    Article  CAS  PubMed  Google Scholar 

  52. Zmuda, F. & Chamberlain, L. H. Regulatory effects of post-translational modifications on zDHHC S-acyltransferases. J. Biol. Chem. 295, 14640–14652 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Lin, L. et al. The Hippo effector YAP promotes resistance to RAF- and MEK-targeted cancer therapies. Nat. Genet. 47, 250–256 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Yun, M. R. et al. Targeting YAP to overcome acquired resistance to ALK inhibitors in ALK-rearranged lung cancer. EMBO Mol. Med. 11, e10581 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Chaib, I. et al. Co-activation of STAT3 and YES-associated protein 1 (YAP1) pathway in EGFR-mutant NSCLC. J. Natl Cancer Inst. https://doi.org/10.1093/jnci/djx014 (2017).

  56. Stephens, R. et al. The Scribble cell polarity module in the regulation of cell signaling in tissue development and tumorigenesis. J. Mol. Biol. 430, 3585–3612 (2018).

    Article  CAS  PubMed  Google Scholar 

  57. Feigin, M. E. et al. Mislocalization of the cell polarity protein scribble promotes mammary tumorigenesis and is associated with basal breast cancer. Cancer Res. 74, 3180–3194 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Shibue, T. & Weinberg, R. A. EMT, CSCs, and drug resistance: the mechanistic link and clinical implications. Nat. Rev. Clin. Oncol. 14, 611–629 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Koo, J. H. & Guan, K. L. Interplay between YAP/TAZ and metabolism. Cell Metab. 28, 196–206 (2018).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This study is supported by the Fund for the Promotion of Joint International Research from JSPS (15KK0303), P-CREATE from the AMED under grant no. 20cm0106513h0005, Princess Takamatsu Cancer Research Fund and Takeda Science Foundation, Aichi Cancer Center Joint Research Project on Priority Areas (to H.E.) and The Hori Sciences and Arts Foundation (to Y.A.). The super-computing resource was provided by Human Genome Center, Institute of Medical Science, The University of Tokyo (http://sc.hgc.jp/shirokane.html).

Author information

Authors and Affiliations

Authors

Contributions

Y.A. and H.E. conceived and designed the project. Y.A. performed most of the experiments. R.K., K.H., Y.N. and S.Y. helped with biochemistry experiments. N.K. helped with animal experiments. R.Y. analyzed the sequencing data. Y.A. and H.E. wrote the manuscript and all authors contributed to the writing and/or critical review of the manuscript.

Corresponding author

Correspondence to Hiromichi Ebi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cancer thanks Sandra Misale and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Scrib mis-localization following KRAS G12C inhibition.

a. The NCI-H358 KRAS G12C mutant lung cancer cell line was treated with 500 nM adagrasib for 48 hours. Representative immunofluorescence images of E-cadherin are shown. Scale bar, 10 μm. b. NCI-H358 cells were treated with 1 μM sotorasib at the indicated time points. Representative immunofluorescence images of Scrib are merged with 4’,6-diamidino-2-phenylindole (DAPI). Scale bar, 5 μm. c. NCI-H358 and LU65 cells were treated with 1 μM sotorasib at the indicated time points. Whole cell extracts (WCE) from untreated cells and cell membrane fractions prepared from each timepoint were blotted with anti-Scrib antibody. CD71 and GAPDH are membrane and cytoplasmic markers, respectively. Western blot was repeated two independent times with similar results. Representative images are shown. d. LU65 cells were transfected with siRNA targeting KRAS or scramble siRNA and cultured for 72 hours. Cells were stained with anti-Scrib or DAPI (blue). Scale bar, 10μm. e. NCI-H358 and LU65 cells were treated with 1 μM sotorasib for 48 or 72 hours and analyzed by FACS to quantify annexin positive cells. The average ± SD of 3 independent experiments is shown. Also, LU65 cells were treated with 1 μM sotorasib for 72 hours or with 5 μM docetaxel for 24 hours. Cells were stained with anti-Scrib (alexa 568), anti-Cleaved PARP (alexa 647), CFSE, a cell permeant molecule used for the fluorescent intracellular labeling of live cells, or DAPI (blue). Scale bar, 10μm. Docetaxel was used as a positive control for induction of cleaved PARP. f. qPCR analysis of mRNA levels of snail and ZEB1 normalized to ubiquitin in NCI-H358 and LU65 cells treated with sotorasib at 1 μM for 48 hours. Data are mean ± SD, n = 3 independently treated cell cultures. g. NCI-H358 single-cell clones with CRISPR-Cas9 KO of Scrib, and introduction of wild-type, P305L mutant, or CAAX-fused Scrib were generated. Each clone and parental cells were lysed and blotted with anti-Scrib antibody. GAPDH is a loading control. h, i. NCI-H358 and LU65 single-cell clones with CRISPR-Cas9 KO of Scrib, or introduction of wild-type or P305L mutant Scrib, were treated with 1 μM sotorasib for 72 hours. Relative cell growth levels were normalized to DMSO-treated cells. All data are represented as mean ± SD, n = 3, independently treated cell cultures (two-sided Student’s t-test, * p < 0.01). j. NCI-H358 or LU65 cells were treated with 1 μM sotorasib for 72 hours. Half of the cells then continued to receive sotorasib with drug replenishment, while treatment was discontinued for the other half. After 72 hours, these cells were re-challenged with sotorasib for 72 hours. Error bars represent mean ± SD, n = 3, independently treated cell cultures. a, b, d, e. Images of immunofluorescence are representative of two independent experiments.

Source data

Extended Data Fig. 2 Inhibition of the driver protein induces Scrib mis-localization in KRAS G12C mutant colorectal cancer, but this is not observed in PI3K-driven breast cancer.

a. NCI-H358 cells expressing cDNA encoding activated AKT (myristoylated AKT (NCI-H358 Myr-AKT)) or MEK (NCI-H358 MEK-DD) were treated with 1 μM sotorasib for 48 hours. Cell membrane fractions were blotted with anti-Scrib and anti-CD71 antibodies. Western blot was repeated two independent times with similar results. Representative images are shown. b. MCF7 (PIK3CA E545K mutant) and T47D (PIK3CA amplified) PI3K-driven breast cancer cell lines were treated with a pan-PI3K inhibitor GDC-0941 at 250 nM for 48 hours and localization of Scrib was determined. c. SW837 and SNU1411 KRAS G12C mutant colorectal cancer cells were treated with 1 μM sotorasib for 48 hours. Representative immunofluorescence images of Scrib are merged with DAPI. Scale bar, 10μm. b, c. Images of immunofluorescence are representative of two independent experiments.

Source data

Extended Data Fig. 3 ZDHHC7 is involved in the regulation of Scrib localization.

a. LU65 cells were treated with 1 μM sotorasib for 48 hours. Representative immunofluorescence images of ZDHHC7 and GM130, a marker for the Golgi apparatus, are shown. Scale bar, 10 μm. b. NCI-H358 cells were treated with 1 μM sotorasib for 48 hours. ZDHHC7 (green) and GM130 (red) were stained and merged. Scale bar, 10μm. The line indicates the profiles taken for the relative fluorescent intensities for ZDHHC7 and GM130. Peaks of fluorescence were identical between ZDHHC7 and GM130. c. The expression of ZDHHC7 in whole cell extracts (WCE), cytoplasm, and nuclear extracts (nucleus) was detected by western blot analysis. Lamin B1 and GAPDH are controls for nuclear and cytoplasmic protein, respectively. d. NCI-H358 and LU65 cells were treated with 1 μM sotorasib for the indicated time points, and lysates were probed with the indicated antibodies. e. qPCR analysis of mRNA levels of ZDHHC7 normalized to ubiquitin in NCI-H358 and LU65 cells treated with 1 μM sotorasib for the indicated time points. Data are mean ± SD, n = 3 independently treated cell cultures. f. NCI-H358 cells were treated with 250 nM GDC-0941 for 48 hours. Representative immunofluorescence images of ZDHHC7 and GM130 are shown. Scale bar, 10μm. g. LU65 cells expressing Myr-AKT or MEK-DD were treated with 1 μM sotorasib for 48 hours. Note that constitutive MEK introduction upregulated ZDHHC7 expression. h. HCC827 and NCI-H2228 cells were treated with 50 nM trametinib for 48 hours. Representative immunofluorescence images of ZDHHC7 and GM130 are shown. Scale bar, 10μm. i. NCI-H358 cells transduced with either control (VC) or ZDHHC7 expression plasmids (OE) were treated with 1 μM sotorasib for 48 hours. Cell membrane fractions were blotted with anti-Scrib and anti-CD71 antibodies, and WCE were blotted with ZDHHC7 and GAPDH. j. NCI-H358 cells were treated with 1 μM sotorasib, 5 μM ML349, or the combination of these drugs for 24 hours. Cell membrane fractions were blotted with anti-Scrib and anti-CD71 antibodies. k. NCI-H358 and LU65 cells were treated with sotorasib, ML349, or the combination of these drugs at the indicated concentrations, and viable cells were measured after 72 hours of treatment. Error bars represent mean ± SD, n = 6, independently treated cell cultures. l. NCI-H358 or LU65 single-cell clones with CRISPR-Cas9 KO of Scrib, and introduction of CAAX-fused Scrib were treated with 1 μM sotorasib with or without 5 μM ML349 for 3 days. Relative cell growth levels were normalized to DMSO-treated cells. All data are represented as mean ± SD n = 3, independently treated cell cultures. a, b, f, h. Images of immunofluorescence are representative of two independent experiments. c, d, g, i, j. Western blot was repeated two independent times with similar results. Representative images are shown.

Extended Data Fig. 4 Scrib mis-localization induces nuclear translocation of YAP.

a, b. NCI-H358 or LU65 single-cell clones with CRISPR-Cas9 KO of Scrib, and introduction of wild-type or P305L mutant Scrib, were stained with Scrib (red), YAP (green), and DAPI, and the images were merged. Fluorescence imaging data are representative of three independent biological experiments. Scale bar, 10 μm. c. NCI-H358 and LU65 cells were treated with 500 nM adagrasib for 48 hours and stained as in a. Images of immunofluorescence are representative of two independent experiments. d. NCI-H358 and LU65 cells were treated with 1 μM sotorasib for the indicated time points, and lysates were probed with the indicated antibodies. e. Lysates from NCI-H358 and LU65 cells transduced with control sgRNA or sgYAP/TAZ were probed with the indicated antibodies. f. g. Optical density readings OD590 nm derived from 10% acetic acid elution of the crystal violet staining from Fig. 4d (for f) and 4e (for g). h. NCI-H358 cells were treated with 1 μM sotorasib, 5 μM ML349, or the combination of these drugs for 48 hours. Cells were stained as in a. Scale bar, 5μm. Images of immunofluorescence are representative of two independent experiments. i. In NCI-H358 and LU65 cell lines, single-cell clones with CRISPR-Cas9 KO of Scrib, and introduction of wild-type or CAAX-fused Scrib were treated with 1 μM sotorasib for the indicated time points and lysates were probed with the indicated antibodies. j. NCI-H358 and LU65 cells were treated with 1 μM sotorasib with or without 5 μM ML349 for the indicated time points and lysates were probed with the indicated antibodies. d, e, i, j. Western blot was repeated two independent times with similar results. Representative images are shown.

Extended Data Fig. 5 Scrib mis-localization induces nuclear translocation of YAP in oncogene-addicted lung cancer models.

a. HCC827 and NCI-H2228 cancer cell lines were treated with 100 nM osimertinib, 500 nM alectinib, or 50 nM trametinib for 72 hours as indicated. Representative immunofluorescence images of Scrib (red) and YAP (green) were merged. Scale bar, 10 μm. Images of immunofluorescence are representative of two independent experiments. b. HCC827 and NCI-H2228 cells were treated with 100 nM osimertinib, or 500 nM alectinib with or without 5 μM ML349 for the indicated time points and lysates were probed with the indicated antibodies. Western blot was repeated two independent times with similar results. Representative images are shown.

Extended Data Fig. 6 The combination of Sotorasib with K-975, a TEAD inhibitor, enhances the efficacy of the KRAS G12C inhibitor.

a. NCI-H358 cells were treated with either DMSO, 1 μM sotorasib, 1μM K-975, or the combination of these drugs for 48 hours. qPCR analysis of mRNA levels of CYR61 and ANKRD1 normalized to ubiquitin following treatment is shown. Data are mean ± SD (n = 3, independently treated cell cultures). b. Optical density readings (OD590 nm) derived from 10% acetic acid elution of the crystal violet staining from Fig. 4g. c. NCI-H358 and LU65 cells were treated with sotorasib, K-975, or the combination of these drugs at the indicated concentrations, and viable cells were measured after 72 hours of treatment. Error bars represent mean ± SD, n = 6, independently treated cell cultures. d. Xenograft tumors derived from NCI-H358 cells were treated as shown in Fig. 4h. These xenografted tumors were harvested at the time of last treatment (Day 24 for vehicle, 38 for K-975, and 66 for sotorasib and the combination of these drugs). mRNA was extracted from these tumors, and qPCR analysis of mRNA levels of CTGF and ANKRD1 are shown. Data are mean ± SD of six tumors. Statistical significance was determined by two-sided Mann–Whitney U Tests. * p = 0.026 for CTGF and <0.01 for ANKRD1, respectively.

Extended Data Fig. 7 Pathways enriched in constitutively active YAP expressed or sotorasib-treated cells.

To analyze the enrichment of the genes upregulated in YAP-S6A expressing cells (n = 3874), sotorasib 72 hr treated cells (n = 3863), and their overlap (n = 845) in NCI-H358, the WEB-based GEne SeT AnaLysis Toolkit (WebGestalt) (http://www.webgestalt.org/) was used. The enrichment of the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and Reactome pathway were processed through Overrepresentation Enrichment Analysis (ORA) with a false discovery rate (FDR) less than 0.05 selected as the cut-off value.

Extended Data Fig. 8 MRAS induction following inhibition of driver oncogenes.

a. NCI-H358 single-cell clones with CRISPR-Cas9 KO of Scrib, and introduction of wild-type or CAAX-fused Scrib, were treated with 1 μM sotorasib for 48 hours and mRNA levels of MRAS were determined by qPCR. Data are mean ± SD (n = 3, independently treated cell cultures). b. NCI-H358 cells were treated with 1 μM sotorasib with or without 5 μM ML349 for 48 hours, and mRNA levels of MRAS normalized to ubiquitin were determined by qPCR. Data are mean ± SD (n = 3, independently treated cell cultures). c. qPCR analysis of mRNA levels of RRAS, RRAS2, and MRAS normalized to ubiquitin in NCI-H358 cells treated with sotorasib at 1 μM for the indicated time points. Data are mean ± SD (n = 3, independently treated cell cultures). d. NCI-H358 and LU65 cells transduced with either control (Ctr) or constitutively active YAP mutant (YAP-S6A) were lysed and blotted with the indicated antibodies. e. NCI-H358 and LU65 cells were treated with either 1 μM sotorasib or 500 nM adagrasib for 72 hours. Representative immunofluorescence images of MRAS are shown. Scale bar, 10 μm. Images of immunofluorescence are representative of three independent experiments. f. HCC827 and NCI-H2228 cells were treated with 100 nM osimertinib or 500 nM alectinib for the indicated time points. Cell lysates were blotted with the indicated antibodies. g. Primer design for ChIP–qPCR. Consensus sequence (GGAATG) recognized and bound by YAP through TEAD around the transcription start site (TSS) are shown as gray vertical bars. Exons are represented as blue vertical bars. Arrows are primers used for ChIP–qPCR in Fig. 5f. d, f. Western blot was repeated two independent times with similar results. Representative images are shown.

Extended Data Fig. 9 MRAS is a cause of feedback activation of the MAPK pathway following sotorasib treatment.

a. qPCR analysis of mRNA levels of MRAS, NRAS, and HRAS normalized to ubiquitin in NCI-H358 cells treated with 1 μM sotorasib for the indicated time points. Data are mean ± SD (n = 3 independently treated cell cultures). b. NCI-H358 cells were treated with 1 μM sotorasib for 4 hours. Scrib (red) and CRAF (green) were stained and merged. Scale bar, 10μm. The lines indicate the profiles taken for the relative fluorescent intensities. While peaks of fluorescence were identical between Scribble and CRAF in untreated cells, the peaks were not merged in sotorasib-treated cells. Images of immunofluorescence are representative of two independent experiments. c. Chemiluminescent quantifications of normalized p-ERK levels from western blots as in Fig. 6d are illustrated. d. NCI-H358 single-cell clones with CRISPR-Cas9 KO of Scrib, and introduction of wild-type or CAAX-fused Scrib, were treated with 1 μM sotorasib for the indicated time points, and lysates were probed with the indicated antibodies. e. Control or YAP/TAZ-knockout cells were treated with 1 μM sotorasib for the indicated time points, and lysates were probed with the indicated antibodies. f. NCI-H358 and LU65 cells were treated with 1 μM sotorasib with or without 5 μM ML349 for the indicated time points. Lysates were probed with the indicated antibodies. GAPDH is a loading control. g. NCI-H358 cells were transfected with either scramble (Scr), NRAS (siNRAS), or MRAS (siMRAS) siRNA and cultured for 24 hours. Media was then replaced, and cells were treated with either 1 μM sotorasib or DMSO for an additional 72 hours. Lysates were probed with the indicated antibodies (left), or relative cell survival was determined (right). Data are mean ± SD, n = 3, independently treated cell cultures. d, e, f, g. Western blot was repeated two independent times with similar results. Representative images are shown.

Extended Data Fig. 10 Tight correlation between EMT markers and MRAS expression.

Scatter plot analysis showing the relationship between MRAS, Vimentin, and ZEB1. Expression data of KRAS mutant lung cancer in TCGA dataset was downloaded from cBioportal. p < 0.0001, both by linear regression analysis.

Supplementary information

Reporting Summary

Supplementary Table

Supplementary Tables 1–3.

Source data

Source Data Fig. 1

Unprocessed western blots for all main figures.

Source Data Fig. 2

Source Data for main figures.

Source Data Extended Data Fig. 1

Source Data for Extended Data Figs.

Source Data Extended Data Fig. 2

Unprocessed blots for Extended Data Figs.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Adachi, Y., Kimura, R., Hirade, K. et al. Scribble mis-localization induces adaptive resistance to KRAS G12C inhibitors through feedback activation of MAPK signaling mediated by YAP-induced MRAS. Nat Cancer 4, 829–843 (2023). https://doi.org/10.1038/s43018-023-00575-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-023-00575-2

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer