Skip to main content

Advertisement

Log in

Ligand-based discovery of small molecules suppressing cancer cell proliferation via autophagic flux inhibition

  • Original Article
  • Published:
Journal of Molecular Medicine Aims and scope Submit manuscript

Abstract

Autophagy is a conserved self-degradation system closely related to cancer progression. Small molecule inhibitors of autophagy have proven to be efficient tools in cancer therapy and are in high demand. Here we report the discovery of two compounds (LZ02/01) capable of suppressing cancer cell proliferation via inhibiting autophagy flux and promoting apoptosis. Potential autophagy inhibitors were selected based on the pharmacophore model derived from the structures of known autophagy inhibitors. LZ02/01-mediated autophagy flux disruption and apoptosis promotion in breast and hepatocellular carcinoma cells (MCF-7 and Hep3B) were examined using a combination of molecular methods in vitro and in vivo. The synergistic tumor-suppressing effects of LZ02 and chloroquine were validated by adopting a xenograft mice model of human breast cancer. Two potential inhibitors (LZ02/01) targeting an autophagy pathway were discovered from the Enamine database. In both MCF-7 and Hep3B cells, LZ02 and LZ01 had the effect of causing the co-occurrence of autophagic flux inhibition and apoptosis induction, robustly suppressing the growth, proliferation, and cell cycle progression. Further tests revealed that FoxO3a and its downstream target genes regulating autophagy, apoptosis, and cell cycle progression were activated and overexpressed, suggesting such effects of LZ02/01 on autophagy and apoptosis were associated with the activation and overexpression of FoxO3a. In addition, LZ02/01-mediated apoptosis is not independent; it was verified to be promoted by autophagic flux inhibition. Meanwhile, synergistic effects on tumor growth reduction were detected in the xenograft mice model of human breast cancer simultaneously treated with LZ02 and chloroquine. Our findings suggest that LZ01 and LZ02 are potent in suppressing cancer cell proliferation and tumor growth through autophagic flux inhibition and apoptosis promotion. The synergistic anti-cancer effects of LZ02 with chloroquine may provide a rational basis for prospective cancer therapy.

Key messages

  • A ligand-based pharmacophore model of high quality is constructed to query hits and two novel scaffold lead compounds LZ01/02 were identified by high-throughput virtual screening.

  • LZ01/02 works to inhibit autophagic flux by attenuating lysosome function.

  • LZ01/02 induces apoptosis through autophagic flux inhibition and apoptosis is the main mechanism to inhibit MCF-7 and Hep3B cancer cell proliferation.

  • The synergistic antitumor growth effects of LZ02 and chloroquine are verified in human xenograft model.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Scheme 1
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9

Similar content being viewed by others

Data availability

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

Atg7:

Autophagy-related gene 7

Bcl-2:

B cell lymphoma 2

Bax:

Bcl2-associated X protein

Bim:

Bcl2 interacting mediator of cell death

CQ:

chloroquine

CTSB:

Cathepsin B

CTSD:

Cathepsin D

LC3:

light chain 3

ULK1:

Unc-51-like kinase 1

References

  1. Liang XH, Jackson S, Seaman M, Brown K, Kempkes B, Hibshoosh H, Levine B (1999) Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature 402:672–676

    CAS  Google Scholar 

  2. Rybstein MD, Bravo-San Pedro JM, Kroemer G, Galluzzi L (2018) The autophagic network and cancer. Nat Cell Biol 20:243–251

    CAS  Google Scholar 

  3. Galluzzi L, Pietrocola F, Levine B, Kroemer G (2014) Metabolic control of autophagy. Cell 159:1263–1276

    CAS  Google Scholar 

  4. Galluzzi L, Baehrecke EH, Ballabio A, Boya P, Bravo-San Pedro JM, Cecconi F, Choi AM, Chu CT, Codogno P, Colombo MI, Cuervo AM, Debnath J, Deretic V, Dikic I, Eskelinen EL, Fimia GM, Fulda S, Gewirtz DA, Green DR, Hansen M, Harper JW, Jäättelä M, Johansen T, Juhasz G, Kimmelman AC, Kraft C, Ktistakis NT, Kumar S, Levine B, Lopez-Otin C, Madeo F, Martens S, Martinez J, Melendez A, Mizushima N, Münz C, Murphy LO, Penninger JM, Piacentini M, Reggiori F, Rubinsztein DC, Ryan KM, Santambrogio L, Scorrano L, Simon AK, Simon HU, Simonsen A, Tavernarakis N, Tooze SA, Yoshimori T, Yuan J, Yue Z, Zhong Q, Kroemer G (2017) Molecular definitions of autophagy and related processes. EMBO J 36:1811–1836

    CAS  Google Scholar 

  5. Karsli-Uzunbas G, Guo JY, Price S, Teng X, Laddha SV, Khor S, Kalaany NY, Jacks T, Chan CS, Rabinowitz JD, White E (2014) Autophagy is required for glucose homeostasis and lung tumor maintenance. Cancer Discov 4:914–927

    CAS  Google Scholar 

  6. Amaravadi R, Debnath J (2014) Mouse models address key concerns regarding autophagy inhibition in cancer therapy. Cancer Discov 4:873–875

    Google Scholar 

  7. Amaravadi RK, Yu D, Lum JJ, Bui T, Christophorou MA, Evan GI, Thomas-Tikhonenko A, Thompson CB (2007) Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. J Clin Invest 117:326–336

    CAS  Google Scholar 

  8. Mariño G, Salvador-Montoliu N, Fueyo A, Knecht E, Mizushima N, López-Otín C (2007) Tissue-specific autophagy alterations and increased tumorigenesis in mice deficient in Atg4C/autophagin-3. J Biol Chem 282:18573–18583

    Google Scholar 

  9. Yue Z, Jin S, Yang C, Levine AJ, Heintz N (2003) Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc Natl Acad Sci 100:15077–15082

    CAS  Google Scholar 

  10. Levine B (2007) Cell biology: autophagy and cancer. Nature 446:745–747

    CAS  Google Scholar 

  11. Mathew R, Karantza-Wadsworth V, White E (2007) Role of autophagy in cancer. Nat Rev Cancer 7:961–967

    CAS  Google Scholar 

  12. Yoshida GJ (2017) Therapeutic strategies of drug repositioning targeting autophagy to induce cancer cell death: from pathophysiology to treatment. J Hematol Oncol 10:67

    Google Scholar 

  13. Egan DF, Chun MG, Vamos M, Zou HX, Rong J, Miller CJ, Lou HJ, Raveendra-Panickar D, Yang C, Sheffler DJ, Teriete P, Asara JM, Turk BE, Cosford ND, Shaw RJ (2015) Small molecule inhibition of the autophagy kinase ULK1 and identification of ULK1 substrates. Mol Cell 59:285–297

  14. Chaikuad A, Koschade SE, Stolz A, Zivkovic K, Pohl C, Shaid S, Ren H, Lambert LJ, Cosford NDP, Brandts CH, Knapp S (2019) Conservation of structure, function and inhibitor binding in UNC-51-like kinase 1 and 2 (ULK1/2). Biochem J 476:875–887

    CAS  Google Scholar 

  15. Limpert AS, Lambert LJ, Bakas NA, Bata N, Brun SN, Shaw RJ, Cosford NDP (2018) Autophagy in cancer: regulation by small molecules. Trends Pharmacol Sci 39:1021–1032

    CAS  Google Scholar 

  16. Deng S, Shanmugam MK, Kumar AP, Yap CT, Sethi G, Bishayee A (2019) Targeting autophagy using natural compounds for cancer prevention and therapy. Cancer

  17. Kurdi A, Cleenewerck M, Vangestel C, Lyssens S, Declercq W, Timmermans J-P, Stroobants S, Augustyns K, de Meyer GRY, van der Veken P, Martinet W (2017) ATG4B inhibitors with a benzotropolone core structure block autophagy and augment efficiency of chemotherapy in mice. Biochem Pharmacol 138:150–162

    CAS  Google Scholar 

  18. Lazarus MB, Shokat KM (2015) Discovery and structure of a new inhibitor scaffold of the autophagy initiating kinase ULK1. Bioorg Med Chem 23:5483–5488

    CAS  Google Scholar 

  19. Lazarus MB, Novotny CJ, Shokat KM (2015) Structure of the human autophagy initiating kinase ULK1 in complex with potent inhibitors. ACS Chem Biol 10:257–261

    CAS  Google Scholar 

  20. Hawkins PC, Skillman AG, Warren GL, Ellingson BA, Stahl MT (2010) Conformer generation with OMEGA: algorithm and validation using high quality structures from the Protein Databank and Cambridge Structural Database. J Chem Inf Model 50:572–584

    CAS  Google Scholar 

  21. Hawkins PC, Nicholls A (2012) Conformer generation with OMEGA: learning from the data set and the analysis of failures. J Chem Inf Model 52:2919–2936

    CAS  Google Scholar 

  22. Wolber G, Langer T (2005) LigandScout: 3-D pharmacophores derived from protein-bound ligands and their use as virtual screening filters. J Chem Inf Model 45:160–169

    CAS  Google Scholar 

  23. Huang N, Shoichet BK, Irwin JJ (2006) Benchmarking sets for molecular docking. J Med Chem 49:6789–6801

    CAS  Google Scholar 

  24. Mysinger MM, Carchia M, Irwin JJ, Shoichet BK (2012) Directory of useful decoys, enhanced (DUD-E): better ligands and decoys for better benchmarking. J Med Chem 55:6582–6594

    CAS  Google Scholar 

  25. Triballeau N, Acher F, Brabet I, Pin JP, Bertrand HO (2005) Virtual screening workflow development guided by the “receiver operating characteristic” curve approach. Application to high-throughput docking on metabotropic glutamate receptor subtype 4. J Med Chem 48:2534–2547

  26. Krautscheid Y, Senning CJÅ, Sartori SB, Singewald N, Schuster D, Stuppner H (2014) Pharmacophore modeling, virtual screening, and in vitro testing reveal haloperidol, eprazinone, and fenbutrazate as neurokinin receptors ligands. J Chem Inf Model 54:1747–1757

    CAS  Google Scholar 

  27. Liu J, Tian Z, Zhou N, Liu X, Liao C, Lei B, Li J, Zhang S, Chen H (2017) Targeting the apoptotic Mcl-1-PUMA interface with a dual-acting compound. Oncotarget 8:54236–54242

    Google Scholar 

  28. Zhao Y, Liu P, Xin Z, Shi C, Bai Y, Sun X, Zhao Y, Wang X, Liu L, Zhao X, Chen Z, Zhang H (2019) Biological characteristics of severe combined immunodeficient mice produced by CRISPR/Cas9-mediated Rag2 and IL2rg mutation. Front Genet 10:401

    CAS  Google Scholar 

  29. de Sabando AR, Wang C, He Y, García-Barros M, Kim J, Shroyer KR, Bannister TD, Yang VW, Bialkowska AB (2016) ML264, a novel small-molecule compound that potently inhibits growth of colorectal cancer. Mol Cancer Ther 15:72–83

  30. Jänicke RU (2009) MCF-7 breast carcinoma cells do not express caspase-3. Breast Cancer Res Treat 117:219–221

    Google Scholar 

  31. Zanella F, Rosado A, García B, Carnero A, Link W (2008) Chemical genetic analysis of FOXO nuclear–cytoplasmic shuttling by using image-based cell screening. Chembiochem 9:2229–2237

    CAS  Google Scholar 

  32. Liu Y, Ao X, Ding W, Ponnusamy M, Wu W, Hao X, Yu W, Wang Y, Li P, Wang J (2018) Critical role of FOXO3a in carcinogenesis. Mol Cancer 17:104

    Google Scholar 

  33. Zachari M, Ganley IG (2017) The mammalian ULK1 complex and autophagy initiation. Essays Biochem 61:585–596

    Google Scholar 

  34. Petherick KJ, Conway OJ, Mpamhanga C, Osborne SA, Kamal A, Saxty B, Ganley IG (2015) Pharmacological inhibition of ULK1 kinase blocks mammalian target of rapamycin (mTOR)-dependent autophagy. J Biol Chem 290:11376–11383

  35. White E, DiPaola RS (2009) The double-edged sword of autophagy modulation in cancer. Clin Cancer Res 15:5308–5316

    Google Scholar 

  36. Wang X, Wei S, Zhao Y, Shi C, Liu P, Zhang C, Lei Y, Zhang B, Bai B, Huang Y, Zhang H (2017) Anti-proliferation of breast cancer cells with itraconazole: hedgehog pathway inhibition induces apoptosis and autophagic cell death. Cancer Lett 385:128–136

    CAS  Google Scholar 

  37. Wang N, Pan W, Zhu M, Zhang M, Hao X, Liang G, Feng Y (2011) Fangchinoline induces autophagic cell death via p53/sestrin2/AMPK signalling in human hepatocellular carcinoma cells. Br J Pharmacol 164:731–742

    CAS  Google Scholar 

  38. Aliwaini S, Swarts AJ, Blanckenberg A, Mapolie S, Prince S (2013) A novel binuclear palladacycle complex inhibits melanoma growth in vitro and in vivo through apoptosis and autophagy. Biochem Pharmacol 86:1650–1663

    CAS  Google Scholar 

  39. Pasquier B (2015) SAR405, a PIK3C3/Vps34 inhibitor that prevents autophagy and synergizes with MTOR inhibition in tumor cells. Autophagy 11:725–726

    CAS  Google Scholar 

  40. El-Khoury V, Pierson S, Szwarcbart E, Brons N, Roland O, Cherrier-De Wilde S, Plawny L, Dyck E, Berchem G (2014) Disruption of autophagy by the histone deacetylase inhibitor MGCD0103 and its therapeutic implication in B-cell chronic lymphocytic leukemia. Leukemia 28:1636–1646

  41. Akin D, Wang SK, Habibzadegah-Tari P, Law B, Ostrov D, Li M, Yin XM, Kim JS, Horenstein N, Dunn WA Jr (2014) A novel ATG4B antagonist inhibits autophagy and has a negative impact on osteosarcoma tumors. Autophagy 10:2021–2035

    Google Scholar 

  42. Doherty J, Baehrecke EH (2018) Life, death and autophagy. Nat Cell Biol 20:1110–1117

    CAS  Google Scholar 

  43. Singh BN, Kumar D, Shankar S, Srivastava RK (2012) Rottlerin induces autophagy which leads to apoptotic cell death through inhibition of PI3K/Akt/mTOR pathway in human pancreatic cancer stem cells. Biochem Pharmacol 84:1154–1163

    CAS  Google Scholar 

  44. Jiang Q, Rao X, Kim CY, Freiser H, Zhang Q, Jiang Z, Li G (2012) Gamma-tocotrienol induces apoptosis and autophagy in prostate cancer cells by increasing intracellular dihydrosphingosine and dihydroceramide. Int J Cancer 130:685–693

    CAS  Google Scholar 

  45. Heidari N, Hicks M, Harada H (2010) GX15-070 (obatoclax) overcomes glucocorticoid resistance in acute lymphoblastic leukemia through induction of apoptosis and autophagy. Cell Death Dis 1:e76

    CAS  Google Scholar 

  46. Zhang Y, Gong J, Xing T, Zheng S, Ding W (2013) Autophagy protein p62/SQSTM1 is involved in HAMLET-induced cell death by modulating apotosis in U87MG cells. Cell Death Dis 4:e550

    CAS  Google Scholar 

  47. Sironi J, Aranda E, Nordstrøm LU, Schwartz EL (2019) Lysosome membrane permeabilization and disruption of the molecular target of rapamycin (mTOR)-lysosome interaction are associated with the inhibition of lung cancer cell proliferation by a chloroquinoline analog. Mol Pharmacol 95:127–138

    CAS  Google Scholar 

  48. Seo SU, Woo SM, Lee HS, Kim SH,Min KJ, Kwon TK (2018) mTORC1/2 inhibitor and curcumin induce apoptosis through lysosomal membrane permeabilization-mediated autophagy. Oncogene 37:5205–5220

  49. Zhao YG, Zhang H (2019) Autophagosome maturation: an epic journey from the ER to lysosomes. J Cell Biol 218:757–770

    CAS  Google Scholar 

  50. Wang F, Tang J, Li P, Si S, Yu H, Yang X, Tao J, Lv Q, Gu M, Yang H, Wang Z (2018) Chloroquine enhances the radiosensitivity of bladder cancer cells by inhibiting autophagy and activating apoptosis. Cell Physiol Biochem 45:54–66

    CAS  Google Scholar 

  51. Golden EB, Cho HY, Hofman FM, Louie SG, Schönthal AH, Chen TC (2015) Quinoline-based antimalarial drugs: a novel class of autophagy inhibitors. Neurosurg Focus 38:E12

  52. Yuan N, Song L, Zhang S, Lin W, Cao Y, Xu F, Fang Y, Wang Z, Zhang H, Li X, Wang Z, Cai J, Wang J, Zhang Y, Mao X, Zhao W, Hu S, Chen S, Wang J (2015) Bafilomycin A1 targets both autophagy and apoptosis pathways in pediatric B-cell acute lymphoblastic leukemia. Haematologica 100:345–356

    CAS  Google Scholar 

  53. Zou Y, Tsai WB, Cheng CJ, Hsu C, Chung YM, Li PC, Lin SH, Hu MCT (2008) Forkhead box transcription factor FOXO3a suppresses estrogen-dependent breast cancer cell proliferation and tumorigenesis. Breast Cancer Res 10:R21

  54. Lin H, Dai T, Xiong H, Zhao X, Chen X, Yu C, Li J, Wang X, Song L (2010) Unregulated miR-96 induces cell proliferation in human breast cancer by downregulating transcriptional factor FOXO3a. PLoS One 5:e15797

    CAS  Google Scholar 

Download references

Acknowledgments

We are grateful to Prof. J.R. Schrock (Emporia State University, USA) for revising the manuscript. We thank Professor Wenxing Ding from University of Kansas Medical Center for his constructive opinions in writing and optimizing the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Li Liu and Zhen Tian contributed equally to this work; Jiyuan Liu, Hai Zhang, and Zhinan Chen conceived the project and designed the experiment; Jiyuan Liu and Yalin Zhang discovered the chemical structures of LZ01 and LZ02 through ligand-based high throughout virtual screening; Jiyuan Liu and Yalin Zhang synthesized LZ01 and LZ02; Jiyuan Liu and Hai Zhang performed the phenotypic screening of LZ01 and LZ 02 on cancer cells and discovered the inhibitory effects of LZ01 and LZ02 on MCF-7 and Hep3B cells; Li Liu, Zhen Tian, and Yifan Li performed the in vitro tests of LZ01 and LZ02 on MCF-7 and Hep3B cells; Peijuan Liu, Zhiqian Xin, Yong Zhao, and Shan Miao carried out the animal experiments; Hai Zhang and Jiyuan Liu prepared the manuscript; Hai Zhang, Jiyuan Liu, and Zhinan Chen supervised the study and contributed reagents/materials; All authors contributed to data analysis.

Corresponding authors

Correspondence to Junling Shi, Zhinan Chen, Jiyuan Liu or Hai Zhang.

Ethics declarations

Competing interests

The authors declare that they have no conflict of interest.

Ethics approval and consent to participate

The experimental protocol for animal studies was reviewed and approved by the Institutional Animal Care and Use Committee (IACUC) of the Fourth Military Medical University.

Consent for publication

Not applicable.

Additional information

Publisher’s note

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

Electronic supplementary material

ESM 1

(DOCX 1587 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, L., Tian, Z., Zhang, Y. et al. Ligand-based discovery of small molecules suppressing cancer cell proliferation via autophagic flux inhibition. J Mol Med 98, 1573–1589 (2020). https://doi.org/10.1007/s00109-020-01971-2

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00109-020-01971-2

Keywords

Navigation