We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Thiosemicarbazones reprogram pancreatic cancer bidirectional oncogenic signaling between cancer cells and stellate cells to suppress desmoplasia

    DR Richardson

    *Author for correspondence:

    E-mail Address: d.richardson@griffith.edu.au

    Centre for Cancer Cell Biology & Drug Discovery, Griffith Institute of Drug Discovery, Griffith University & School of Environment & Science (N34), Nathan, Brisbane, Queensland, 4111, Australia

    Department of Pathology & Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466–8550, Japan

    ,
    M Gholam Azad

    Centre for Cancer Cell Biology & Drug Discovery, Griffith Institute of Drug Discovery, Griffith University & School of Environment & Science (N34), Nathan, Brisbane, Queensland, 4111, Australia

    ,
    R Afroz

    Centre for Cancer Cell Biology & Drug Discovery, Griffith Institute of Drug Discovery, Griffith University & School of Environment & Science (N34), Nathan, Brisbane, Queensland, 4111, Australia

    ,
    V Richardson

    Centre for Cancer Cell Biology & Drug Discovery, Griffith Institute of Drug Discovery, Griffith University & School of Environment & Science (N34), Nathan, Brisbane, Queensland, 4111, Australia

    &
    M Dharmasivam

    Centre for Cancer Cell Biology & Drug Discovery, Griffith Institute of Drug Discovery, Griffith University & School of Environment & Science (N34), Nathan, Brisbane, Queensland, 4111, Australia

    Published Online:https://doi.org/10.4155/fmc-2022-0050

    Standard treatments have shown dismal activity against pancreatic cancer (PC), due in part to the development of a dense stroma (desmoplasia). This perspective discusses the development of the di-2-pyridylketone thiosemicarbazones that overcomes bidirectional oncogenic signaling between PC cells and pancreatic stellate cells (PSCs), which is critical for desmoplasia development. This activity is induced by the up-regulation of the metastasis suppressor, N-myc downstream-regulated gene-1 (NDRG1), which inhibits oncogenic signaling via HGF, IGF-1 and Sonic Hedgehog pathway. More recent studies have deciphered additional pathways including those mediated by Wnt and tenascin C that are secreted by PSCs to activate β-catenin and YAP/TAZ signaling in PC cells. Suppression of bidirectional signaling between cell types presents a unique therapeutic opportunity.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Rahib L, Smith BD, Aizenberg R et al. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 74(11), 2913–2921 (2014).
    • 2. Neuzillet C, Tijeras-Raballand A, Bourget P et al. State of the art and future directions of pancreatic ductal adenocarcinoma therapy. Pharmacol. Ther. 155, 80–104 (2015).
    • 3. Chand S, O'Hayer K, Blanco FF et al. The landscape of pancreatic cancer therapeutic resistance mechanisms. Int. J. Biol. Sci. 12(3), 273–282 (2016).
    • 4. Sagol Ö, Yavuzsen T, Öztop I et al. The effect of apoptotic activity, survivin, Ki-67, and P-glycoprotein expression on prognosis in pancreatic carcinoma. Pancreas. 30(4), 343–348 (2005).
    • 5. O'Driscoll L, Walsh N, Larkin A et al. MDR1/P-glycoprotein and MRP-1 drug efflux pumps in pancreatic carcinoma. Anticancer Res. 27(4B), 2115–2120 (2007).
    • 6. Neesse A, Michl P, Frese KK et al. Stromal biology and therapy in pancreatic cancer. Gut. 60(6), 861–868 (2011).
    • 7. Vonlaufen A, Joshi S, Qu C et al. Pancreatic stellate cells: partners in crime with pancreatic cancer cells. Cancer Res. 68(7), 2085–2093 (2008).
    • 8. Dauer P, Nomura A, Saluja A, Banerjee S. Microenvironment in determining chemo-resistance in pancreatic cancer: neighborhood matters. Pancreatology. 17(1), 7–12 (2017).
    • 9. Creeden JF, Alganem K, Imami AS et al. Kinome array profiling of patient-derived pancreatic ductal adenocarcinoma identifies differentially active protein tyrosine kinases. Int. J. Mol. Sci. 21(22), 8679 (2020).
    • 10. Creeden JF, Alganem K, Imami AS et al. Emerging kinase therapeutic targets in pancreatic ductal adenocarcinoma and pancreatic cancer desmoplasia. Int. J. Mol. Sci. 21(22), 8823 (2020).
    • 11. Hassouneh B, Islam M, Nagel T et al. Tetrathiomolybdate promotes tumor necrosis and prevents distant metastases by suppressing angiogenesis in head and neck cancer. Mol. Cancer Ther. 6(3), 1039–1045 (2007).
    • 12. Kalinowski DS, Stefani C, Toyokuni S et al. Redox cycling metals: pedaling their roles in metabolism and their use in the development of novel therapeutics. Biochim. Biophys. Acta, Mol. Cell Res. 1863(4), 727–748 (2016).
    • 13. Ishida S, Andreux P, Poitry-Yamate C, Auwerx J, Hanahan D. Bioavailable copper modulates oxidative phosphorylation and growth of tumors. Proc. Natl. Acad. Sci. 110(48), 19507–19512 (2013).
    • 14. Lener MR, Scott RJ, Wiechowska-Kozłowska A et al. Serum concentrations of selenium and copper in patients diagnosed with pancreatic cancer. Cancer Res. Treat. Official J. Korean Cancer Association. 48(3), 1056 (2016).
    • 15. Kalousová M, Krechler T, Jáchymová M et al. Ferritin as an independent mortality predictor in patients with pancreas cancer. Results of a pilot study. Tumor Biol. 33(5), 1695–1700 (2012).
    • 16. Toshiyama R, Konno M, Eguchi H et al. Association of iron metabolic enzyme hepcidin expression levels with the prognosis of patients with pancreatic cancer. Oncol. Lett. 15(5), 8125–8133 (2018).
    • 17. Kalinowski DS, Richardson DR. The evolution of iron chelators for the treatment of iron overload disease and cancer. Pharmacol. Rev. 57(4), 547–583 (2005).
    • 18. Merlot AM, Kalinowski DS, Richardson DR. Novel chelators for cancer treatment: where are we now? Antioxid. Redox Signal. 18(8), 973–1006 (2013).
    • 19. Richardson D, Tran E, Ponka P. The potential of iron chelators of the pyridoxal isonicotinoyl hydrazone class as effective antiproliferative agents. Blood. 86, 4295–4306 (1995).
    • 20. Lovejoy DB, Richardson DR. Novel ‘hybrid’ iron chelators derived from aroylhydrazones and thiosemicarbazones demonstrate selective antiproliferative activity against tumor cells. Blood. 100(2), 666–676 (2002).
    • 21. Richardson DR, Sharpe PC, Lovejoy DB et al. Dipyridyl thiosemicarbazone chelators with potent and selective antitumor activity form iron complexes with redox activity. J. Med. Chem. 49(22), 6510–6521 (2006).
    • 22. Whitnall M, Howard J, Ponka P, Richardson DR. A class of iron chelators with a wide spectrum of potent antitumor activity that overcomes resistance to chemotherapeutics. Proc. Natl. Acad. Sci. 103(40), 14901–14906 (2006).
    • 23. Yuan J, Lovejoy DB, Richardson DR. Novel di-2-pyridyl–derived iron chelators with marked and selective antitumor activity: in vitro and in vivo assessment. Blood. 104(5), 1450–1458 (2004).
    • 24. Kovacevic Z, Chikhani S, Lovejoy DB, Richardson DR. Novel thiosemicarbazone iron chelators induce up-regulation and phosphorylation of the metastasis suppressor N-myc down-stream regulated gene 1: a new strategy for the treatment of pancreatic cancer. Mol. Pharmacol. 80(4), 598–609 (2011).
    • 25. Liu W, Xing F, Iiizumi-Gairani M et al. N-myc downstream regulated gene 1 modulates Wnt-β-catenin signalling and pleiotropically suppresses metastasis. EMBO Mol. Med. 4(2), 93–108 (2012).
    • 26. Yu Y, Rahmanto YS, Richardson D. Bp44mT: an orally active iron chelator of the thiosemicarbazone class with potent anti-tumour efficacy. Br. J. Pharmacol. 165(1), 148–166 (2012).
    • 27. Geleta B, Park KC, Jansson PJ et al. Breaking the cycle: Targeting of NDRG1 to inhibit bi-directional oncogenic cross-talk between pancreatic cancer and stroma. FASEB J. 35(2), e21347 (2021). •• This article is of considerable interest as it provides the evidence regarding the inhibition of bidirectional signaling between pancreatic cancer cells and pancreatic stellate cells.
    • 28. Kovacevic Z, Menezes SV, Sahni S et al. The metastasis suppressor, N-MYC downstream-regulated gene-1 (NDRG1), down-regulates the ErbB family of receptors to inhibit downstream oncogenic signaling pathways. J. Biol. Chem. 291(3), 1029–1052 (2016).
    • 29. Lovejoy DB, Sharp DM, Seebacher N et al. Novel second-generation di-2-pyridylketone thiosemicarbazones show synergism with standard chemotherapeutics and demonstrate potent activity against lung cancer xenografts after oral and intravenous administration in vivo. J. Med. Chem. 55(16), 7230–7244 (2012).
    • 30. Guo ZL, Richardson DR, Kalinowski DS et al. The novel thiosemicarbazone, di-2-pyridylketone 4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC), inhibits neuroblastoma growth in vitro and in vivo via multiple mechanisms. J. Hematol. Oncol. 9(1), 1–16 (2016).
    • 31. Lim SC, Jansson PJ, Assinder SJ et al. Unique targeting of androgen-dependent and-independent AR signaling in prostate cancer to overcome androgen resistance. FASEB J. 34(9), 11511–11528 (2020).
    • 32. Sestak V, Stariat J, Cermanova J et al. Novel and potent anti-tumor and anti-metastatic di-2-pyridylketone thiosemicarbazones demonstrate marked differences in pharmacology between the first and second generation lead agents. Oncotarget. 6(40), 42411 (2015).
    • 33. Jansson PJ, Kalinowski DS, Lane DJ et al. The renaissance of polypharmacology in the development of anti-cancer therapeutics: inhibition of the ‘triad of death’ in cancer by Di-2-pyridylketone thiosemicarbazones. Pharmacol. Res. 100, 255–260 (2015).
    • 34. Lovejoy DB, Jansson PJ, Brunk UT et al. Antitumor activity of metal-chelating compound Dp44mT is mediated by formation of a redox-active copper complex that accumulates in lysosomes. Cancer Res. 71(17), 5871–5880 (2011).
    • 35. Stacy AE, Palanimuthu D, Bernhardt PV et al. Zinc (II)–thiosemicarbazone complexes are localized to the lysosomal compartment where they transmetallate with copper ions to induce cytotoxicity. J. Med. Chem. 59(10), 4965–4984 (2016).
    • 36. Hu M, Carraway KL III. Repurposing cationic amphiphilic drugs and derivatives to engage lysosomal cell death in cancer treatment. Front. Oncol. 2837 (2020).
    • 37. Stefan SM, Jansson PJ, Kalinowski DS et al. The growing evidence for targeting P-glycoprotein in lysosomes to overcome resistance. Future Med. Chem. 12(6), 473–477 (2020).
    • 38. Appelqvist H, Waster P, Kagedal K, Ollinger K. The lysosome: from waste bag to potential therapeutic target. J. Mol. Cell Biol. 5(4), 214–226 (2013).
    • 39. Jansson PJ, Yamagishi T, Arvind A et al. Di-2-pyridylketone 4, 4-dimethyl-3-thiosemicarbazone (Dp44mT) overcomes multidrug resistance by a novel mechanism involving the hijacking of lysosomal P-glycoprotein (Pgp). J. Biol. Chem. 290(15), 9588–9603 (2015).
    • 40. Seebacher NA, Richardson DR, Jansson PJ. A mechanism for overcoming P-glycoprotein-mediated drug resistance: novel combination therapy that releases stored doxorubicin from lysosomes via lysosomal permeabilization using Dp44mT or DpC. Cell Death Dis. 7(12), e2510–e2510 (2016).
    • 41. Yamagishi T, Sahni S, Sharp DM et al. P-glycoprotein mediates drug resistance via a novel mechanism involving lysosomal sequestration. J. Biol. Chem. 288(44), 31761–31771 (2013).
    • 42. Stacy AE, Palanimuthu D, Bernhardt PV et al. Structure–activity relationships of di-2-pyridylketone, 2-benzoylpyridine, and 2-acetylpyridine thiosemicarbazones for overcoming Pgp-mediated drug resistance. J. Med. Chem. 59(18), 8601–8620 (2016).
    • 43. Seebacher NA, Lane DJ, Jansson PJ, Richardson DR. Glucose modulation induces lysosome formation and increases lysosomotropic drug sequestration via the P-glycoprotein drug transporter. J. Biol. Chem. 291(8), 3796–3820 (2016).
    • 44. Le NT, Richardson DR. Iron chelators with high antiproliferative activity up-regulate the expression of a growth inhibitory and metastasis suppressor gene: a link between iron metabolism and proliferation. Blood. 104(9), 2967–2975 (2004).
    • 45. Hosoi F, Izumi H, Kawahara A et al. N-myc downstream regulated gene 1/Cap43 suppresses tumor growth and angiogenesis of pancreatic cancer through attenuation of inhibitor of κB kinase β expression. Cancer Res. 69(12), 4983–4991 (2009).
    • 46. Maruyama Y, Ono M, Kawahara A et al. Tumor growth suppression in pancreatic cancer by a putative metastasis suppressor gene Cap43/NDRG1/Drg-1 through modulation of angiogenesis. Cancer Res. 66(12), 6233–6242 (2006).
    • 47. Kovacevic Z, Chikhani S, Lui GY et al. The iron-regulated metastasis suppressor NDRG1 targets NEDD4L, PTEN, and SMAD4 and inhibits the PI3K and Ras signaling pathways. Antioxid. Redox Signal. 18(8), 874–887 (2013).
    • 48. Menezes SV, Fouani L, Huang ML et al. The metastasis suppressor, NDRG1, attenuates oncogenic TGF-β and NF-κB signaling to enhance membrane E-cadherin expression in pancreatic cancer cells. Carcinogenesis. 40(6), 805–818 (2019).
    • 49. Chen Z, Zhang D, Yue F et al. The iron chelators Dp44mT and DFO inhibit TGF-beta-induced epithelial-mesenchymal transition via up-regulation of N-Myc downstream-regulated gene 1 (NDRG1). J. Biol. Chem. 287(21), 17016–17028 (2012).
    • 50. Jin R, Liu W, Menezes S et al. The metastasis suppressor NDRG1 modulates the phosphorylation and nuclear translocation of β-catenin through mechanisms involving FRAT1 and PAK4. J. Cell Sci. 127(14), 3116–3130 (2014).
    • 51. Liu W, Yue F, Zheng M et al. The proto-oncogene c-Src and its downstream signaling pathways are inhibited by the metastasis suppressor, NDRG1. Oncotarget. 6(11), 8851 (2015).
    • 52. Sun J, Zhang D, Zheng Y et al. Targeting the metastasis suppressor, NDRG1, using novel iron chelators: regulation of stress fiber-mediated tumor cell migration via modulation of the ROCK1/pMLC2 signaling pathway. Mol. Pharmacol. 83(2), 454–469 (2013).
    • 53. Wangpu X, Lu J, Xi R et al. Targeting the metastasis suppressor, N-Myc downstream regulated gene-1, with novel di-2-pyridylketone thiosemicarbazones: Suppression of tumor cell migration and cell-collagen adhesion by inhibiting focal adhesion kinase/paxillin signaling. Mol. Pharmacol. 89(5), 521–540 (2016).
    • 54. Park KC, Geleta B, Leck LYW et al. Thiosemicarbazones suppress expression of the c-Met oncogene by mechanisms involving lysosomal degradation and intracellular shedding. J. Biol. Chem. 295(2), 481–503 (2020).
    • 55. Menezes SV, Kovacevic Z, Richardson DR. The metastasis suppressor NDRG1 down-regulates the epidermal growth factor receptor via a lysosomal mechanism by up-regulating mitogen-inducible gene 6. J. Biol. Chem. 294(11), 4045–4064 (2019).
    • 56. Liby KT, Yore MM, Sporn MB. Triterpenoids and rexinoids as multifunctional agents for the prevention and treatment of cancer. Nat. Rev. Cancer. 7(5), 357–369 (2007).
    • 57. Korcsmáros T, Szalay MS, Böde C et al. How to design multi-target drugs: target search options in cellular networks. Expert Opin. Drug Discov. 2(6), 799–808 (2007).
    • 58. Campbell PJ, Yachida S, Mudie LJ et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature. 467(7319), 1109–1113 (2010).
    • 59. Pruijn FB, Sturman JR, Liyanage HS et al. Extravascular transport of drugs in tumor tissue: effect of lipophilicity on diffusion of tirapazamine analogues in multicellular layer cultures. J. Med. Chem. 48(4), 1079–1087 (2005).
    • 60. Kleeff J, Korc M, Apte M et al. Pancreatic cancer. Nat. Rev. Dis. Primers. 2(1), 1–22 (2016).
    • 61. Apte MV, Park S, Phillips PA et al. Desmoplastic reaction in pancreatic cancer: role of pancreatic stellate cells. Pancreas. 29(3), 179–187 (2004).
    • 62. Pothula SP, Xu Z, Goldstein D et al. Hepatocyte growth factor inhibition: a novel therapeutic approach in pancreatic cancer. Br. J. Cancer. 114(3), 269–280 (2016).
    • 63. Rucki AA, Foley K, Zhang P et al. Heterogeneous stromal signaling within the tumor microenvironment controls the metastasis of pancreatic cancer. Cancer Res. 77(1), 41–52 (2017).
    • 64. Tape CJ, Ling S, Dimitriadi M et al. Oncogenic KRAS regulates tumor cell signaling via stromal reciprocation. Cell. 165(4), 910–920 (2016).
    • 65. Gu D, Schlotman KE, Xie J. Deciphering the role of hedgehog signaling in pancreatic cancer. J. Biomed. Res. 30(5), 353 (2016).
    • 66. Kasperczyk H, Baumann B, Debatin KM, FuMa S. Characterization of sonic hedgehog as a novel NF-κB target gene that promotes NF-κB-mediated apoptosis resistance and tumor growth in vivo. FASEB J. 23(1), 21–33 (2009).
    • 67. Nakashima H, Nakamura M, Yamaguchi H et al. Nuclear factor-κB contributes to hedgehog signaling pathway activation through sonic hedgehog induction in pancreatic cancer. Cancer Res. 66(14), 7041–7049 (2006).
    • 68. Noubissi FK, Goswami S, Sanek NA et al. Wnt signaling stimulates transcriptional outcome of the Hedgehog pathway by stabilizing GLI1 mRNA. Cancer Res. 69(22), 8572–8578 (2009).
    • 69. Adamska A, Domenichini A, Falasca M. Pancreatic Ductal Adenocarcinoma: Current and Evolving Therapies. Int. J. Mol. Sci. 18(7), 1338 (2017).
    • 70. Liu Q, Liao Q, Zhao Y. Chemotherapy and tumor microenvironment of pancreatic cancer. Cancer Cell Int. 17(1), 1–17 (2017).
    • 71. Geleta B, Tout FS, Lim SC et al. Targeting Wnt/Tenascin C-mediated cross talk between pancreatic cancer cells and stellate cells via activation of the metastasis suppressor NDRG1. J. Biol. Chem. 101608 (2022). • Provides additional molecular evidence regarding the ability of NDRG1 to inhibit bidirectional crosstalk between pancreatic cancer cells and pancreatic stellate cells.
    • 72. Pothula SP, Xu Z, Goldstein D et al. Targeting the HGF/c-MET pathway: stromal remodelling in pancreatic cancer. Oncotarget. 8(44), 76722 (2017).
    • 73. Xu Z, Vonlaufen A, Phillips PA et al. Role of pancreatic stellate cells in pancreatic cancer metastasis. Am. J. Pathol. 177(5), 2585–2596 (2010).
    • 74. Apte MV, Pirola RC, Wilson JS. Pancreatic stellate cells: a starring role in normal and diseased pancreas. Front. Physiol. 3, 344 (2012).
    • 75. Fujita H, Ohuchida K, Mizumoto K et al. α-Smooth muscle actin expressing stroma promotes an aggressive tumor biology in pancreatic ductal adenocarcinoma. Pancreas. 39(8), 1254–1262 (2010).