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:

Therapeutic targeting of the mevalonate–geranylgeranyl diphosphate pathway with statins overcomes chemotherapy resistance in small cell lung cancer

Abstract

Small cell lung cancer (SCLC) lacks effective treatments to overcome chemoresistance. Here we established multiple human chemoresistant xenograft models through long-term intermittent chemotherapy, mimicking clinically relevant therapeutic settings. We show that chemoresistant SCLC undergoes metabolic reprogramming relying on the mevalonate (MVA)–geranylgeranyl diphosphate (GGPP) pathway, which can be targeted using clinically approved statins. Mechanistically, statins induce oxidative stress accumulation and apoptosis through the GGPP synthase 1 (GGPS1)–RAB7A–autophagy axis. Statin treatment overcomes both intrinsic and acquired SCLC chemoresistance in vivo across different SCLC PDX models bearing high GGPS1 levels. Moreover, we show that GGPS1 expression is negatively associated with survival in patients with SCLC. Finally, we demonstrate that combined statin and chemotherapy treatment resulted in durable responses in three patients with SCLC who relapsed from first-line chemotherapy. Collectively, these data uncover the MVA–GGPP pathway as a metabolic vulnerability in SCLC and identify statins as a potentially effective treatment to overcome chemoresistance.

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: Establishment of chemoresistant SCLC xenograft mouse models.
Fig. 2: Statins suppress chemoresistant SCLC cells by reduction of MVA.
Fig. 3: Statins effectively inhibit chemoresistant SCLC growth.
Fig. 4: Statins function through the GGPS1–RAB7A–autophagy axis.
Fig. 5: Statins overcome acquired chemoresistance in GGPS1high SCLC.
Fig. 6: Statins overcome intrinsic chemoresistance in GGPS1high SCLC.
Fig. 7: Statins plus chemotherapy overcome chemoresistance in SCLC.

Similar content being viewed by others

Data availability

RNA-seq data generated have been deposited in the Gene Expression Omnibus under accession no. GSE160241. The 112 human SCLC RNA expression data were derived from the TCGA Research Network: (http://cancergenome.nih.gov/). The dataset derived from this resource that supports the findings of this study is available as follows: 81 specimens from database no. EGAS00001000925 (European Genome–Phenome Archive under the accession code) and 31 specimens from database no. EGAS00001000334 (EBI)6. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding author upon reasonable request.

Code availability

No new algorithms were developed for this manuscript.

References

  1. Siegel, R. L., Miller, K. D. & Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 70, 7–30 (2020).

    Article  PubMed  Google Scholar 

  2. Sabari, J. K., Lok, B. H., Laird, J. H., Poirier, J. T. & Rudin, C. M. Unravelling the biology of SCLC: implications for therapy. Nat. Rev. Clin. Oncol. 14, 549–561 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Yu, J. B., Decker, R. H., Detterbeck, F. C. & Wilson, L. D. Surveillance epidemiology and end results evaluation of the role of surgery for stage I small cell lung cancer. J. Thorac. Oncol. 5, 215–219 (2010).

    Article  PubMed  Google Scholar 

  4. Gazdar, A. F., Bunn, P. A. & Minna, J. D. Small-cell lung cancer: what we know, what we need to know and the path forward. Nat. Rev. Cancer 17, 725–737 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. Poirier, J. T. et al. New approaches to SCLC therapy: from the laboratory to the clinic. J. Thor. Oncol. 15, 520–540 (2020).

    Article  Google Scholar 

  6. George, J. et al. Comprehensive genomic profiles of small cell lung cancer. Nature 524, 47–53 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Gardner, E. E. et al. Chemosensitive relapse in small cell lung cancer proceeds through an EZH2-SLFN11 axis. Cancer Cell 31, 286–299 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Drapkin, B. J. et al. Genomic and functional fidelity of small cell lung cancer patient-derived xenografts. Cancer Discov. 8, 600–615 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Stewart, C. A. et al. Single-cell analyses reveal increased intratumoral heterogeneity after the onset of therapy resistance in small-cell lung cancer. Nat. Cancer 1, 423–436 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Keibler, M. A. et al. Metabolic requirements for cancer cell proliferation. Cancer Metab. 4, 16 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Vander Heiden, M. G. & DeBerardinis, R. J. Understanding the intersections between metabolism and cancer biology. Cell 168, 657–669 (2017).

    Article  PubMed Central  CAS  Google Scholar 

  12. Longo, J. et al. An actionable sterol-regulated feedback loop modulates statin sensitivity in prostate cancer. Mol. Metab. 25, 119–130 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Goldstein, J. L. & Brown, M. S. Regulation of the mevalonate pathway. Nature 343, 425–430 (1990).

    Article  CAS  PubMed  Google Scholar 

  14. Clendening, J. W. & Penn, L. Z. Targeting tumor cell metabolism with statins. Oncogene 31, 4967–4978 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. Garcia-Bermudez, J. et al. Squalene accumulation in cholesterol auxotrophic lymphomas prevents oxidative cell death. Nature 567, 118–122 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Xia, Z. et al. Blocking protein geranylgeranylation is essential for lovastatin-induced apoptosis of human acute myeloid leukemia cells. Leukemia 15, 1398–1407 (2001).

    Article  CAS  PubMed  Google Scholar 

  17. Demierre, M. F., Higgins, P. D., Gruber, S. B., Hawk, E. & Lippman, S. M. Statins and cancer prevention. Nat. Rev. Cancer 5, 930–942 (2005).

    Article  CAS  PubMed  Google Scholar 

  18. Moon, S. H. et al. p53 Represses the mevalonate pathway to mediate tumor suppression. Cell 176, 564–580 (2019).

    Article  CAS  PubMed  Google Scholar 

  19. Cristea, S. et al. The MEK5-ERK5 kinase axis controls lipid metabolism in small-cell lung cancer. Cancer Res. 80, 1293–1303 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Augert, A. et al. MAX functions as a tumor suppressor and rewires metabolism in small cell lung cancer. Cancer Cell 38, 97–114 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Morita, M. et al. PKM1 confers metabolic advantages and promotes cell-autonomous tumor cell growth. Cancer Cell 33, 355–367 (2018).

    Article  CAS  PubMed  Google Scholar 

  22. Huang, F. et al. Inosine monophosphate dehydrogenase dependence in a subset of small cell lung cancers. Cell Metab. 28, 369–382 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 16, 582–598 (2016).

    Article  CAS  PubMed  Google Scholar 

  24. Corsello, S. M. et al. Discovering the anti-cancer potential of non-oncology drugs by systematic viability profiling. Nat. Cancer 1, 235–248 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Fendt, S. M., Frezza, C. & Erez, A. Targeting metabolic plasticity and flexibility dynamics for cancer therapy. Cancer Discov. 10, 1797–1807 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Rao, S. et al. Lovastatin-mediated G1 arrest is through inhibition of the proteasome, independent of hydroxymethyl glutaryl-CoA reductase. Proc. Natl Acad. Sci. USA 96, 7797–7802 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Clendening, J. W. et al. Dysregulation of the mevalonate pathway promotes transformation. Proc. Natl Acad. Sci. USA 107, 15051–15056 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wu, J. et al. Intercellular interaction dictates cancer cell ferroptosis via NF2-YAP signalling. Nature 572, 402–406 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Xia, Y. et al. The mevalonate pathway is a druggable target for vaccine adjuvant discovery. Cell 175, 1059–1073 (2018).

    Article  CAS  PubMed  Google Scholar 

  30. Seabra, M. C. & Wasmeier, C. Controlling the location and activation of Rab GTPases. Curr. Opin. Cell Biol. 16, 451–457 (2004).

    Article  CAS  PubMed  Google Scholar 

  31. Mancuso, M., Orsucci, D., Volpi, L., Calsolaro, V. & Siciliano, G. Coenzyme Q10 in neuromuscular and neurodegenerative disorders. Curr. Drug Targets 11, 111–121 (2010).

    Article  CAS  PubMed  Google Scholar 

  32. Wong, W. W., Dimitroulakos, J., Minden, M. D. & Penn, L. Z. HMG-CoA reductase inhibitors and the malignant cell: the statin family of drugs as triggers of tumor-specific apoptosis. Leukemia 16, 508–519 (2002).

    Article  CAS  PubMed  Google Scholar 

  33. Mullen, P. J., Yu, R., Longo, J., Archer, M. C. & Penn, L. Z. The interplay between cell signalling and the mevalonate pathway in cancer. Nat. Rev. Cancer 16, 718–731 (2016).

    Article  CAS  PubMed  Google Scholar 

  34. Longo, J., van Leeuwen, J. E., Elbaz, M., Branchard, E. & Penn, L. Z. Statins as anticancer agents in the era of precision medicine. Clin. Cancer Res. 26, 5791–5800 (2020).

    Article  CAS  PubMed  Google Scholar 

  35. Stenmark, H. Rab GTPases as coordinators of vesicle traffic. Nat. Rev. Mol. Cell Biol. 10, 513–525 (2009).

    Article  CAS  PubMed  Google Scholar 

  36. Zinn, R. L. et al. Combination treatment with ABT-737 and chloroquine in preclinical models of small cell lung cancer. Mol Cancer 12, 16 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Lin, Z. et al. Mevastatin blockade of autolysosome maturation stimulates LBH589-induced cell death in triple-negative breast cancer cells. Oncotarget 8, 17833–17848 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Li, L., Tan, J., Miao, Y., Lei, P. & Zhang, Q. ROS and autophagy: interactions and molecular regulatory mechanisms. Cell. Mol. Neurobiol. 35, 615–621 (2015).

    Article  PubMed  CAS  Google Scholar 

  39. Gao, L., Loveless, J., Shay, C. & Teng, Y. Targeting ROS-mediated crosstalk between autophagy and apoptosis in cancer. Adv. Exp. Med. Biol. 1260, 1–12 (2020).

    Article  CAS  PubMed  Google Scholar 

  40. McGregor, G. H. et al. Targeting the metabolic response to statin-mediated oxidative stress produces a synergistic antitumor response. Cancer Res. 80, 175–188 (2020).

    Article  CAS  PubMed  Google Scholar 

  41. Schulze, A. & Harris, A. L. How cancer metabolism is tuned for proliferation and vulnerable to disruption. Nature 491, 364–373 (2012).

    Article  CAS  PubMed  Google Scholar 

  42. Pandyra, A. et al. Immediate utility of two approved agents to target both the metabolic mevalonate pathway and its restorative feedback loop. Cancer Res. 74, 4772–4782 (2014).

    Article  CAS  PubMed  Google Scholar 

  43. Nakao, M. et al. Nab-paclitaxel monotherapy for relapsed small cell lung cancer: retrospective analysis and review. Anticancer Res. 40, 1579–1585 (2020).

    Article  CAS  PubMed  Google Scholar 

  44. Gelsomino, F. et al. Phase 2 study of NAB-paclitaxel in SensiTivE and refractory relapsed small cell lung cancer (SCLC) (NABSTER TRIAL). Br. J. Cancer 123, 26–32 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Hodgkinson, C. L. et al. Tumorigenicity and genetic profiling of circulating tumor cells in small-cell lung cancer. Nat. Med. 20, 897–903 (2014).

    Article  CAS  PubMed  Google Scholar 

  46. Lim, J. S. et al. Intratumoural heterogeneity generated by Notch signalling promotes small-cell lung cancer. Nature 545, 360–364 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Christensen, C. L. et al. Targeting transcriptional addictions in small cell lung cancer with a covalent CDK7 inhibitor. Cancer Cell 26, 909–922 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Mollaoglu, G. et al. MYC drives progression of small cell lung cancer to a variant neuroendocrine subtype with vulnerability to aurora kinase inhibition. Cancer Cell 31, 270–285 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Jia, D. et al. Crebbp loss drives small cell lung cancer and increases sensitivity to HDAC inhibition. Cancer Discov. 8, 1422–1437 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Grunblatt, E. et al. MYCN drives chemoresistance in small cell lung cancer while USP7 inhibition can restore chemosensitivity. Genes Dev. 34, 1210–1226 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhang, H. et al. CDK7 inhibition potentiates genome instability triggering anti-tumor immunity in small cell lung cancer. Cancer Cell 37, 37–54 (2020).

    Article  CAS  PubMed  Google Scholar 

  52. Iams, W. T., Porter, J. & Horn, L. Immunotherapeutic approaches for small-cell lung cancer. Nat. Rev. Clin. Oncol. 17, 300–312 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Paz-Ares, L. et al. Durvalumab plus platinum-etoposide versus platinum-etoposide in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): a randomised, controlled, open-label, phase 3 trial. Lancet 394, 1929–1939 (2019).

    Article  CAS  PubMed  Google Scholar 

  54. Kim, J. et al. CPS1 maintains pyrimidine pools and DNA synthesis in KRAS/LKB1-mutant lung cancer cells. Nature 546, 168–172 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Kim, H. S. et al. Systematic identification of molecular subtype-selective vulnerabilities in non-small-cell lung cancer. Cell 155, 552–566 (2013).

    Article  CAS  PubMed  Google Scholar 

  56. Seo, Y. K. et al. Genome-wide localization of SREBP-2 in hepatic chromatin predicts a role in autophagy. Cell Metab. 13, 367–375 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Khanzada, U. K. et al. Potent inhibition of small-cell lung cancer cell growth by simvastatin reveals selective functions of Ras isoforms in growth factor signalling. Oncogene 25, 877–887 (2006).

    Article  CAS  PubMed  Google Scholar 

  58. Mahoney, C. E. et al. A chemical biology screen identifies a vulnerability of neuroendocrine cancer cells to SQLE inhibition. Nat. Commun. 10, 96 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Seckl, M. J. et al. Multicenter, phase III, randomized, double-blind, placebo-controlled trial of pravastatin added to first-line standard chemotherapy in small-cell lung cancer (LUNGSTAR). J. Clin. Oncol. 35, 1506–1514 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Han, J. Y. et al. A phase 2 study of irinotecan, cisplatin, and simvastatin for untreated extensive-disease small cell lung cancer. Cancer 117, 2178–2185 (2011).

    Article  CAS  PubMed  Google Scholar 

  61. Wiederschain, D. et al. Single-vector inducible lentiviral RNAi system for oncology target validation. Cell Cycle 8, 498–504 (2009).

    Article  CAS  PubMed  Google Scholar 

  62. Nemati, F. et al. Distinctive potentiating effects of cisplatin and/or ifosfamide combined with etoposide in human small cell lung carcinoma xenografts. Clin. Cancer Res. 6, 2075–2086 (2000).

    CAS  PubMed  Google Scholar 

  63. Zhao, G. et al. Cullin5 deficiency promotes small-cell lung cancer metastasis by stabilizing integrin beta1. J. Clin. Invest. 129, 972–987 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Li, F. et al. LKB1 inactivation elicits a redox imbalance to modulate non-small cell lung cancer plasticity and therapeutic response. Cancer Cell 27, 698–711 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Ireland, A. S. et al. MYC drives temporal evolution of small cell lung cancer subtypes by reprogramming neuroendocrine fate. Cancer Cell 38, 60–78 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Zhou, C. et al. Monitoring autophagic flux by an improved tandem fluorescent-tagged LC3 (mTagRFP-mWasabi-LC3) reveals that high-dose rapamycin impairs autophagic flux in cancer cells. Autophagy 8, 1215–1226 (2012).

    Article  CAS  PubMed  Google Scholar 

  67. Chhonker, Y. S., Haney, S. L., Bala, V., Holstein, S. A. & Murry, D. J. Simultaneous quantitation of isoprenoid pyrophosphates in plasma and cancer cells using LC-MS/MS. Molecules 23, 3275 (2018).

  68. Jemal, M., Schuster, A. & Whigan, D. B. Liquid chromatography/tandem mass spectrometry methods for quantitation of mevalonic acid in human plasma and urine: method validation, demonstration of using a surrogate analyte, and demonstration of unacceptable matrix effect in spite of use of a stable isotope analog internal standard. Rapid Commun. Mass Spectrom. 17, 1723–1734 (2003).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Natural Science Foundation of China (grant nos. 82030083 to H.J. and 82173340 and 81871875 to L.H.); the National Basic Research Program of China (grant nos. 2017YFA0505501 and 2020YFA0803300 to H.J.); the Strategic Priority Research Program of the Chinese Academy of Sciences (grant no. XDB19020201 to H.J.); the National Natural Science Foundation of China (grant nos. 81872312, 82011540007 and 31621003 to H.J., 91957120 to S.L., 81402371 to Y.J., 81802279 to H.H. and 81902326 to X.W.); the Basic Frontier Scientific Research Program of Chinese Academy of Science (no. ZDBS-LY-SM006 to H.J.); the International Cooperation Project of Chinese Academy of Sciences (no. 153D31KYSB20190035 to H.J.); Science and Technology Commission of Shanghai Municipality (no. 21ZR1470300 to L.H.); and the Youth Innovation Promotion Association CAS (no. Y919S31371 to X.W.). R.K.T. is supported by the German Research Foundation Deutsche Forschungsgemeinsaft as part of SFB1399 (grant no. 413326622) and by the German Ministry of Science and Education as part of the e:Med program (InCa, grant no. 01ZX1901A). We thank W. Wei, L. Tan, Y. Feng, H. Zhang, F. Li, Y. Gao, X. Liu, C. Xu, D. Gao, L. Zhang and X. He for helpful comments, and X. Jiang, Y. Zhang, Z. Liu and L. Huang for material sharing.

Author information

Authors and Affiliations

Authors

Contributions

H.J., L.H. and C.G. conceived the idea and designed the experiments. C.G., R.W. and Y.H. performed all experiments and analyzed data. S.-H.L., Y.Q., T.Z., Q.Z., Y.N., Y.J., H.-Y.H., X.W., L.T., R.K.T., H.Z. and K.-K.W. provided technical assistance and contributed intellectually. Y.H. provided human SCLC biopsy specimens. J.C. and L.C. performed bioinformatics analyses. C.G., L.H. and H.J. wrote the manuscript.

Corresponding authors

Correspondence to Liang Hu or Hongbin Ji.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cancer thanks the anonymous reviewers 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 Establishment of chemoresistant SCLC xenograft mouse models.

(a) Representative IHC staining and quantification of γH2AX staining in H82 and H82R tumors treated with or without E/P (n = 9 tissue sections from three mice). (b and c) Representative IHC staining and quantification of CC3 (b) and γH2AX staining (c) in H209 and H209R tumors treated with or without E/P (n = 9 tissue sections from three mice). (d and e) Representative IHC staining and quantification of CC3 (d) and γH2AX staining (e) in H526 and H526R tumors treated with or without E/P (n = 9 tissue sections from three mice). (f and g) Representative IHC staining and quantification of CC3 (f) and γH2AX staining (g) in H146 and H146R tumors treated with or without E/P (n = 9 tissue sections from three mice). (h) Relative viability of H82 and H82R cells treated upon 0.1μM DDP with indicated concentrations of VP16 for 72 hrs (n = 3 cell culture replicates). Data were shown as means. One of three independent experiments was shown. (i) Relative viability of H526 and H526R cells treated with E/P (0.25 μM VP16 & 0.1μM DDP) for 96 hrs (n = 3 cell culture replicates). One of three independent experiments was shown. (j and k) Representative IHC staining and quantification of CC3 (j) and γH2AX staining (k) in H446 tumors treated with or without E/P (n = 9 tissue sections from three mice). (l and m) Representative IHC staining and quantification of CC3 (l) and γH2AX staining (m) in DMS114 tumors treated with or without E/P (n = 9 tissue sections from three mice). (n) Relative viability of H196 cells treated with E/P (0.25 μM VP16 & 0.1μM DDP) in vitro for 96 hrs (n = 3 cell culture replicates). One of three independent experiments was shown. Scale bars: 50 μm (a-g, j-m). Data were shown as mean ± S.E.M (a-g, j-m). Statistical significance was calculated by two-tailed unpaired Student’s t-test (a-g).

Source data

Extended Data Fig. 2 Statin suppresses chemoresistant SCLC growth via reducing MVA.

(a) Top 10 candidates (red) preferentially inhibiting H82R cell viability in comparison to H82 cells (H82R/H82) from the screening of FDA-approved 1971 drug library. Pravastatin (blue) was also shown. (b) Distribution of the putative targets of 256 FDA-approved metabolic drugs categorized by human diseases. (c) Distribution of the putative targets of 256 FDA-approved drugs categorized by metabolism pathways. (d) Work flow of 256 FDA-approved metabolic drug screening. H82R and H82 cells were treated with individual drugs (5 μM) and cell viability was measured after 72 hrs treatment. (e) Volcano plot showing log2 fold change against log10 P value for each drug in H82 and H82R cells (3 independent cell cultures for each drug). Drugs with the preferential inhibition in H82R cells (-log2 ratio of H82R/H82 > 1 and p < 0.05) were shown in red and those with the preferential inhibition in H82 cells (-log2 ratio of H82R/H82 < -0.5 and p < 0.05) were shown in blue. (f) Relative viability of H82 and H82R cells after 72 hrs treatment with increasing concentrations of pravastatin (n = 3 cell culture replicates). One of two independent experiments was shown. (g) Morphology of H82R cells treated with mevastatin (5 μM) for 72 hrs. (h) Western blotting analysis of cPARP, CC3, P27, P21, and PCNA levels in H82 and H82R cells after treatment with mevastatin (5 μM). (i) Western blotting analysis of HMGCR protein levels in 293 T cells with HMGCR overexpression (left) and H82R cells with shRNA-mediated HMGCR knockdown (right). (j) Western blotting analysis of HMGCR protein levels in H82 and H82R cells. (k) Mass spectrometry (MS) analysis of the relative abundance of IPP/DMAPP, GPP, FPP and GGPP in H82 and H82R cells (n = 4 independent cell cultures). Morphology (g) and western blot (h-j) images were representative of three independent experiments with similar results. Arrow indicated Cleaved PARP (h). Actin served as the internal control. Scale bars: 100 μm (g). Data were shown as mean ± S.E.M (k). Statistical significance was calculated by two-tailed unpaired Student’s t-test (e, k).

Source data

Extended Data Fig. 3 Statin effectively inhibits chemoresistant SCLC growth.

(a and b) Relative viability of 4 chemosensitiveand and 4 chemoresistant cell lines treated with E/P (0.25 μM VP16 & 0.1μM DPP) (a) or mevastatin (5 μM) (b) for 96 hrs (n = 3 cell culture replicates per cell line). See also Fig. 3a. (c) Western blotting analysis of cPARP, CC3, P27, P21, and PCNA levels in chemosensitive cell lines (H209, H526) and chemoresistant cell lines (DMS114, H446) treated with mevastatin (Mev) for 72 hrs. (d and e) Relative viability of H209/H209R (d) and H526/H526R (e) treated with mevastatin (5 μM) for 72 hrs (n = 2 cell culture replicates). One of two independent experiments was shown. (f and g) Tumor (f) and mouse (g) weights from mice treated in Fig. 3b. (n = 7 mice per group). (h) Representative images of Ki-67 IHC staining (left) in H82R tumors treated in Fig. 3b. and quantification of Ki-67 H-Score in H82R tumors (right) treated in Fig. 3b. (n = 9 tissue sections from seven mice). (i) Tumor growth of H82R tumor-bearing mice treated with Control (ctrl) (n = 3 mice), E/P (n = 3 mice), simvastatin (Sim) (n = 2 mice), Sim + E/P (n = 3 mice), pitvastatin (Pit) (n = 2 mice) or Pit + E/P (n = 3 mice). (j and k) Tumor (j) and mouse (k) weights from mice treated in (i). (l and m) Tumor (l) and mouse (m) weights from mice treated in Fig. 3e. (n) Representative images of Ki-67 IHC staining (left) in H446 tumors treated in Fig. 3e and quantification of Ki-67 H-Score in H446 tumors (right) treated in Fig. 3e (n = 9 tissue sections from seven mice). Western blot (c) images were representative of three independent experiments with similar results. Arrow indicated Cleaved PARP (c). Actin served as the internal control. Scale bars: 50 μm (h, n). Data were shown as mean ± S.E.M (f-n). Statistical significance was calculated by one-way ANOVA with Dunnett’s multiple comparisons test (f-n).

Source data

Extended Data Fig. 4 GGPP supplementation reverses the statin’s effects.

(a) Schematic of the MVA pathway. The key enzymes are shown in green and their pharmacological inhibitors are shown in red. The MVA downstream pathways are also indicated. (b to d) Relative viability of H82 and H82R cells treated with increasing concentrations of FDPS inhibitors risedronate, or zoledronate, or TH-Z93 (b), FDFT1 inhibitors BHP-652 or ZA (c), SQLE inhibitor NB-598, SOAT1 inhibitor avasimibe, or NPC1L1 inhibitor ezetimibe (d) for 72 hrs (n = 2 cell culture replicates). One of two independent experiments was shown. (e) The relative cell growth of H82 and H82R cells cultured with or without lipoprotein-depleted serum (LPDS) medium (n = 2 cell culture replicates). (f to j) Relative viability of H82 and H82R cells treated with increasing concentrations of 5-lipoxygenase zileuton (f), GSH synthesis inhibitor erastin (g) or BSO (h), ferroptosis inducer ML-162 or RSL-3 (i), farnesyltransferase inhibitor tipifarnib (j) for 72 hrs (n = 2 cell culture replicates). One of two independent experiments was shown. (k) Measurement of OCR in H82 and H82R cells (n = 5 cell culture replicates). (l) Relative viability of H82 and H82R cells treated with increasing concentrations of GGPS1 inhibitor TH-Z145 for 72 hrs (n = 3 cell culture replicates). One of two independent experiments was shown. (m) Western blotting analysis of GGPS1, SQLE and FDFT1 protein levels in H82 and H82R cells. (n) Western blotting analysis of EZH2 and MYC levels in H82 and H82R cells. (o) Relative viability of H82 and H82R cells treated with increasing concentrations of EZH2 inhibitor JQZ5 and MYC inhibitor BAY1238097 for 72 hrs (n = 3 cell culture replicates). One of two independent experiments was shown. (p) Relative viability of H82R cells co-treated with mevastatin (5 μM) alone or plus increasing concentrations of FPP, FFOH, GGPP, GGOH, SQ, coenzyme Q9, coenzyme Q10, or Idebenone for 72 hrs. FFOH: farnesol, the donor of a farnesyl group. SQ: squalene. CoQ9, Q10: Coenzyme Q9, Q10 (n = 2 cell culture replicates). (q and r) Relative viability of H446 (q) and DMS114 cells (r) co-treated with mevastatin (5 μM) alone or plus individual intermediate metabolites (250 mM MVA, 2 μM FPP, 0.1 μM FFOH, 2 μM GGPP, 10 μM GGOH, 0.1 μM SQ, 10 μM CoQ9, 10 μM CoQ10, 1 μM Idebenone). H446 cells (n = 2 cell culture replicates) were treated with 5 μM mevastatin for 72 hrs and the DMS114 cells (n = 3 cell culture replicates) were treated with 10 μM mevastatin for 96 hrs. One of two independent experiments was shown. (s) MS analysis of the relative abundance of MVA, FPP and GGPP in H82R cells co-treated with mevastatin (5 μM) alone or plus individual intermediate metabolites (250 mM MVA or 2 μM GGPP) for 24 hrs (n = 4 independent cell cultures). (t) MS analysis of the relative abundance of MVA (left) and GGPP (right) in H82R cells treated with pravastatin (20 μM) for 24 hrs (n = 4 independent cell cultures). Western blot images (m-n) were representative of three independent experiments with similar results. Actin served as the internal control. Data were shown as mean ± S.E.M (s-t). Statistical significance was calculated by two-tailed unpaired Student’s t-test (s).

Source data

Extended Data Fig. 5 Statin functions through the GGPS1-RAB7A-autophagy axis.

(a) Mean gene expression of various small RAB GTPases (FPKM) in H82R cells from RNA-seq analysis (n = 3 biological independent samples). (b) Relative cell growth of H82R and H82 cells with Dox-induced knockdown of GGPS1 or RAB7A (n = 3 cell culture replicates). Cells were treated with Dox during the entire experiment. Data were shown as means. One of two independent experiments was shown. (c) Compare the viability of H82R (n = 2 cell culture replicates) and H82 (n = 3 cell culture replicates) cells with Dox-induced knockdown of GGPS1 or RAB7A at day 4. One of two independent experiments was shown. (d) Western blotting analysis of P62 and LC3B protein levels in chemosensitive (H209, H526) and chemoresistant (H446, DMS114) cell lines treated with mevastatin (5 μM) for 72 hrs. (e) Western blotting analysis of cPARP and CC3 levels in H82 and H82R cells treated with mevastatin (5 μM) alone or plus GGPP (2 μM) for 72 hrs. (f) Western blotting analysis of cPARP and CC3 levels in H82R cells with Dox-induced knockdown of GGPS1 or RAB7A. (g) Relative cell growth of H446 cells with or without Dox-induced knockdown of GGPS1 or RAB7A (n = 3 cell culture replicates). Data were shown as means. One of two independent experiments was shown. (h) Western blotting analysis of GGPS1, RAB7A, P62 and LC3B levels in H446 cells with Dox-induced knockdown of GGPS1 or RAB7A. (i) Relative viability of H82 and H82R cells with chloroquine (20 μM) or Baf-A1 (2 nM) treatment (n = 2 cell culture replicates). One of two independent experiments was shown. (j) Representative immunofluorescence images indicating the autophagosome in H446 cells expressing mRFPGFPLC3 fusion protein in response to mevastatin (5 μM) alone or plus GGPP (2 μM) for 72 hrs treatment. (k) Quantification of RFP+GFP+ and RFP+GFP- puncta in H446 cells treated in (j). (n = 19/14/13 independent cells of Ctrl/ Mev/ Mev+GGPP groups respectively). (l and m) Relative viability of 2 chemosensitive (H209, H526) and 2 chemoresistant cells (H446, DMS114) treated with increasing concentrations of FDPS inhibitor TH-Z93 (l) or GGPS1 inhibitor TH-Z145 (m) for 72 hrs (n = 3 cell culture replicates per cell line). (n) Western blotting analysis of P62 and LC3B levels in chemosensitive (H526) and chemoresistant (DMS114) cells treated with TH-Z145 (0.3 mM). (o) Western blotting analysis of P62 and LC3B levels in H82R tumors treated with mevastatin alone or plus E/P. (p) Relative viability of H82R cells pre-incubated with z-VAD (20 μM) or IETD (20 μM) for 2 hrs then treated with mevastatin (10 μM) 24 hrs (n = 3 cell culture replicates). One of three independent experiments was shown. (q) Western blotting analysis of cPARP, CC8 (cleaved caspase 8) and CC3 levels in H82R cells pre-incubated with z-VAD (20 μM) or IETD (20 μM) for 2 hrs then treated with mevastatin (10 μM) 24 hrs. (r) FACS analysis of relative ROS levels in DMS114 and H526 cells after 48 hrs treatment with E/P or mevastatin (5 μM) treatment (n = 3 independent cell cultures). (s) FACS analysis of relative ROS levels in H82R and H82 cells with Dox-induced knockdown of GGPS1 or RAB7A for 48 hrs (n = 3 independent cell cultures). Western blot (d-f, h, n-o, q) and immunofluorescence (j) results were representative of three independent experiments with similar results. Arrow indicated Cleaved LC3B (d, h, n-o) or Cleaved PARP (e-f, q). Tubulin or Actin served as the internal control. Scale bars: 5μm (j). Data were shown as mean ± S.E.M (k, r-s). Statistical significance was calculated by one-way ANOVA with Dunnett’s multiple comparisons test (k, r-s).

Source data

Extended Data Fig. 6 Knockdown of GGPS1 or RAB7A phenocopied the statin’s effects.

(a) Subcutaneous H82R tumors from mice treated in Fig. 4o. (b) Tumor weights from mice treated in Fig. 4o (n = 5 mice per group). (c) Representative IHC staining of GGPS1 and 8-OXO (c) and quantification of GGPS1-positive cells and 8-OXO H-Score (d) in H82R tumors treated in Fig. 4o (n = 9 tissue sections from five mice). (e) Mouse weights from Fig. 4o (n = 5 mice per group). (f) Tumor growth of H82 tumor-bearing mice with or without inducible GGPS1 knockdown (n = 3 mice per group). (g) Tumor weights from mice treated in (f). (h) Representative IHC staining of GGPS1 in H82 tumors treated in (f). (i) Representative IHC staining of CC3 and quantification of CC3 positive cells in H82 tumors treated in (f) (n = 9 tissue sections from three mice). (j) Tumor growth of H82R tumor-bearing mice with or without inducible RAB7A knockdown (n = 5 mice per group). (k) Subcutaneous H82R tumors from mice treated in (j). (l) Tumor weights from mice treated in (j). (m) Representative H&E and IHC staining of RAB7A, CC3, γH2AX and 8-OXO (m) and quantification of RAB7A, CC3, γH2AX positive cells and 8-OXO H-Score (n) in H82R tumors treated in (j) (n = 6 tissue sections from five mice). (o) Western blotting analysis of P62 and LC3B levels in H82R tumors treated in (j). (p) Relative cell growth of H82R cells with Dox treatment (n = 3 cell culture replicates). Cells were treated with Dox during the entire experiment. Data were shown as means. One of two independent experiments was shown. (q) Tumor growth of H82R tumor-bearing mice with Dox treatment (n = 5 mice per group). (r-s) Tumor (r) and mouse (s) weights from mice treated in (q). Immunohistochemistry (h) and western blot (o) results were representative of three independent experiments with similar results. Arrow indicated Cleaved LC3B (o). Tubulin served as the internal control. Scale bars: 50 μm (c, h-i, m). Data were shown as mean ± S.E.M (b, d-g, i-j, l, n, q-s). Statistical significance was calculated by two-tailed unpaired Student’s t-test (b, d, j, l, n).

Source data

Extended Data Fig. 7 Statins overcome acquired chemoresistance in GGPS1high SCLC.

(a) Western blotting analysis of HMGCR, FDPS, FDFT1 and SQLE levels in chemosensitive and chemoresistant cell lines. (b) MS analysis of the relative abundance of IPP/DMAPP (H446, P < 0.0001; DMS114, P < 0.0001; H196, P < 0.0001), GPP (H446, P < 0.0001; DMS114, P = 0.0747; H196, P = 0.4009) and FPP (H446, P < 0.0001; DMS114, P = 0.0014; H196, P < 0.0001) in chemosensitive and chemoresistant cell lines. Data shown as the relative value of these metabolites in comparison to mevalonate (n = 4 independent cell cultures per cell line). (c and d) Representative IHC and quantification of CC3 (c) and γH2AX staining (d) in SC234 and SC234R tumors treated with or without E/P (n = 9 tissue sections from three mice). (e and f) Representative IHC and quantification of CC3 (e) and γH2AX staining (f) in SC224 and SC224R tumors treated with or without E/P (n = 9 tissue sections from three mice). (g to i) Western blotting analysis of HMGCR, FDPS, GGPS1, FDFT1 and SQLE levels in SC234/SC234R tumors (g), SC256/SC256R tumors (h) and SC224/SC224R tumors (i). (j) Tumor weights from mice treated in Fig. 5i (n = 5 mice per group). (k) Representative IHC and quantification of 8-OXO staining SC234R tumors treated in Fig. 5i (n = 9 tissue sections from five mice). (l and m) Tumor (l) and mouse (m) weights from mice treated in Fig. 5l. (n = 3 mice per group). (n) Representative IHC and quantification of 8-OXO staining SC256R tumors treated in Fig. 5l (n = 9 tissue sections from three mice). (o and p) Tumor (o) and mouse (p) weights from mice treated in Fig. 5o (n = 4 mice per group). (q) Representative IHC and quantification of 8-OXO staining SC224R tumors treated in Fig. 5o (n = 9 tissue sections from four mice). Western blot (a, g-i) results were representative of three independent experiments with similar results. Tubulin served as the internal control. Scale bars: 50 μm (c-f, k, n, q). Data were shown as mean ± S.E.M (b-f, j-q). Statistical significance was calculated by two-tailed unpaired Student’s t-test (b-f) or one-way ANOVA with Dunnett’s multiple comparisons test (j-n).

Source data

Extended Data Fig. 8 GGPS1 levels in SCLC PDX tumors with various drug responses.

(a to c) PDX mouse models were established using transbronchoscopic lung biopsy specimens from treatment-naïve SCLC patients and subjected to E/P treatment. Ticks on the X axis indicated day1 of each E/P cycle. These patients had differential responses to chemotherapy in the clinic: SC022, SC007, SC158 and SC185 had PR (partial response) (a); SC073 and SC441 had SD (stable disease) (b); SC009 had PD (progressive disease) (c). (d to e) Representative IHC staining of NCAM (d) and GGPS1 (e) in various SCLC PDX mouse tumors. PR, partial response; SD, stable disease; PD, progressive disease. Scale bars: 50 μm. Immunohistochemistry results (d-e) were representative of three independent experiments with similar results. Scale bars: 50 μm (d-e). Data were shown as mean ± S.E.M (a-c). Statistical significance was calculated by two-tailed unpaired Student’s t-test (a-c).

Source data

Extended Data Fig. 9 Statins overcome intrinsic chemoresistance in GGPS1high SCLC.

(a) Western blotting analysis of HMGCR level in indicated PDX tumors used in Fig. 6b. (b) Subcutaneous SC009 tumors from mice treated in Fig. 6d. (c and d) Tumor (c) and mouse (d) weights from mice treated in Fig. 6d. (n = 5 mice per group). (e) Representative IHC and quantification of 8-OXO staining of SC009 tumors treated in Fig. 6d (n = 9 tissue sections from five mice). (f and g) Tumor (f) and mouse (g) weights from mice treated in Fig. 6g (n = 5 mice per group). (h) Representative IHC and quantification of 8-OXO staining of SC359 tumors treated in Fig. 6g (n = 9 tissue sections from five mice). (i to k) Survival analysis of SCLC patients according to HMGCR (i), SQLE (j) and FDFT1 (k) expression status in TCGA database. (l) Q-PCR analysis of HMGCR and HMGCS1 levels in H82 and H82R cells treated with mevastatin (5 μM) for 16 hrs. (m) Q-PCR analysis of HMGCR and HMGCS1 levels in H446 and DMS114 cells treated with mevastatin (5 μM) for 16 hrs. (n and o) Q-PCR analysis of HMGCR and HMGCS1 levels in SC009 (n) or SC359 (o) tumors treated with mevastatin. Two samples per group with three replicates. (p) Q-PCR analysis of HMGCR and HMGCS1 levels in H209 and H209R cells treated with mevastatin (5 μM) for 16 hrs. (q) Q-PCR analysis of HMGCR and HMGCS levels in H526 and H526R cells treated with mevastatin (5 μM) for 16 hrs. Results (l-m, p-q) were representative of two independent experiments with similar results with three replicates. Scale bars: 50 μm (e, h). Data were shown as mean ± S.E.M (c-h). Statistical significance was calculated by one-way ANOVA with Dunnett’s multiple comparisons test (c-h).

Source data

Extended Data Fig. 10 Statin plus chemotherapy overcomes chemoresistance in SCLC.

(a) Representative IHC staining of GGPS1 in two transbronchoscopic lung biopsy specimens from SCLC patients relapsed form chemotherapy (SP016 and SP017). Representative tumor cells were indicated by the orange arrows. (b) Patient SP983: CT scan images at baseline and after 4 weeks of simvastatin plus chemotherapy (etoposide/carboplatin). The in situ lung tumor was indicated by the orange arrows. (c) Patient SP772: CT and MRI scan images at baseline and after 9 weeks of simvastatin plus chemotherapy (albumin-bound paclitaxel). Orange arrows indicated tumors. Top panel: in situ primary lung tumor; Middle and bottom panels: two intracranial metastatic tumors. The maximum diameters of two intracranial metastatic lesions were decreased from 1.2 cm to 0.4 cm and 1.0 cm to 0.6 cm, respectively. (d) The PFS comparison of patients with chemotherapy plus simvastatin (n = 2 patients; SP651, SP772) and patients with chemotherapy alone (n = 4 patients; SP112, SP235, SP347, SP509). The PFS calculation was based on the CT scanning at baseline and after the changes of treatment regimens. The SP983 patient was not available for long term monitor. (e) The median PFS from patients relapsed from first-line chemotherapy in previous reports. Nakao et al. analyzed 17 relapsed SCLC patients followed by albumin-bound paclitaxel treatment. Gelsomino et al. analyzed 43 relapsed SCLC patients followed by albumin-bound paclitaxel treatment. Gelsomino et al. analyzed 25 refractory SCLC patients followed by albumin-bound paclitaxel treatment. (f) Routine blood test of hemoglobin (HGB), white blood cell (WBC), absolute number of neutrophils (NEUT#) and platelet (PLT) in the three patients who received simvastatin plus chemotherapy. (g) Biochemical index of alanine aminotransferase (ALT), aspartate aminotransferase (AST), total bilirubin (TBIL) and direct bilirubin (DBIL) in the three patients who received simvastatin plus chemotherapy. (h) Biochemical index of urea nitrogen (UREA), creatinine (CREA) in the three patients who received simvastatin plus chemotherapy. (i) Biochemical index of creatine kinase (CK), creatine kinase-MB isoenzyme (CK-MB) in the patients who received simvastatin plus chemotherapy. Immunohistochemistry results (a) were representative of three independent experiments with similar results. Scale bars: 50 μm (a). Data were shown as mean ± S.E.M (d-e). Statistical significance was calculated by two-tailed unpaired Student’s t-test (d).

Source data

Supplementary information

Supplementary Information

Supplementary Fig. 1.

Reporting Summary

Supplementary Tables

Supplementary Tables 1. Screening of 1,971 FDA drugs. 2. Screening of 256 FDA drugs. 3.Primer information. 4.PDX information. 5.Compounds and inhibitors. 6. information on human patients.

Source data

Source Data Fig. 1

Statistical Source data.

Source Data Fig. 2

Statistical Source data.

Source Data Fig. 3

Statistical Source data.

Source Data Fig. 4

Statistical Source data.

Source Data Fig. 4

Unprocessed immunoblots.

Source Data Fig. 5

Statistical Source data.

Source Data Fig. 5

Unprocessed immunoblots.

Source Data Fig. 6

Statistical Source data.

Source Data Fig. 6

Unprocessed immunoblots.

Source Data Extended Data Fig. 1

Statistical Source data.

Source Data Extended Data Fig. 2

Statistical Source data.

Source Data Extended Data Fig. 2

Unprocessed immunoblots.

Source Data Extended Data Fig. 3

Statistical Source data.

Source Data Extended Data Fig. 3

Unprocessed immunoblots.

Source Data Extended Data Fig. 4

Statistical Source data.

Source Data Extended Data Fig. 4

Unprocessed immunoblots.

Source Data Extended Data Fig. 5

Statistical Source data.

Source Data Extended Data Fig. 5

Unprocessed immunoblots.

Source Data Extended Data Fig. 6

Statistical Source data.

Source Data Extended Data Fig. 6

Unprocessed immunoblots.

Source Data Extended Data Fig. 7

Statistical Source data.

Source Data Extended Data Fig. 7

Unprocessed immunoblots.

Source Data Extended Data Fig. 8

Statistical Source data.

Source Data Extended Data Fig. 9

Statistical Source data.

Source Data Extended Data Fig. 9

Unprocessed immunoblots.

Source Data Extended Data Fig. 10

Statistical Source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Guo, C., Wan, R., He, Y. et al. Therapeutic targeting of the mevalonate–geranylgeranyl diphosphate pathway with statins overcomes chemotherapy resistance in small cell lung cancer. Nat Cancer 3, 614–628 (2022). https://doi.org/10.1038/s43018-022-00358-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-022-00358-1

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