Skip to main content
Top
Published in: Investigational New Drugs 3/2020

Open Access 01-06-2020 | Melanoma | PRECLINICAL STUDIES

Anticancer activity of a novel methylated analogue of L-mimosine against an in vitro model of human malignant melanoma

Authors: Sotiris Kyriakou, Melina Mitsiogianni, Theodora Mantso, William Cheung, Stephen Todryk, Stephany Veuger, Aglaia Pappa, David Tetard, Mihalis I. Panayiotidis

Published in: Investigational New Drugs | Issue 3/2020

Login to get access

Summary

The anticancer activity of a series of novel synthesized, hydroxypyridone-based metal chelators (analogues of L-mimosine) was evaluated in an in vitro model of melanoma consisting of malignant melanoma (A375), non-melanoma epidermoid carcinoma (A431) and immortalized non-malignant keratinocyte (HaCaT) cells. More specifically, we have demonstrated that the L-enantiomer of a methylated analogue of L-mimosine (compound 22) can exert a potent anticancer effect in A375 cells when compared to either A431 or HaCaT cells. Moreover, we have demonstrated that this analogue has the ability to i) promote increased generation of reactive oxygen species (ROS), ii) activate both intrinsic and extrinsic apoptosis and iii) induce perturbations in cell cycle growth arrest. Our data highlights the potential of compound 22 to act as a promising therapeutic agent against an in vitro model of human malignant melanoma.
Appendix
Available only for authorised users
Literature
1.
go back to reference Orthaber K, Pristovnik MM, Skok K, Perić B, Maver UU (2017) Skin Cancer and its treatment: novel treatment approaches with emphasis on nanotechnology. J Nanomater 2017:1–20 Orthaber K, Pristovnik MM, Skok K, Perić B, Maver UU (2017) Skin Cancer and its treatment: novel treatment approaches with emphasis on nanotechnology. J Nanomater 2017:1–20
3.
go back to reference Rastrelli M, Tropea S, Rossi CR, Alaibac M (2014) Melanoma: epidemiology, risk factors, pathogenesis, diagnosis and classification. In Vivo 28(6):1005–1011PubMed Rastrelli M, Tropea S, Rossi CR, Alaibac M (2014) Melanoma: epidemiology, risk factors, pathogenesis, diagnosis and classification. In Vivo 28(6):1005–1011PubMed
4.
go back to reference Zbytek B, Carlson JA, Granese J, Ross J, Mihm MC Jr, Slominski A (2008) Current concepts of metastasis in melanoma. Expert Rev Dermatol 3(5):569–585PubMedPubMedCentral Zbytek B, Carlson JA, Granese J, Ross J, Mihm MC Jr, Slominski A (2008) Current concepts of metastasis in melanoma. Expert Rev Dermatol 3(5):569–585PubMedPubMedCentral
5.
go back to reference Gupte A, Mumper RJ (2009) Elevated copper and oxidative stress in Cancer cells as a target for Cancer treatment. Cancer Treat Rev 35(1):32–46PubMed Gupte A, Mumper RJ (2009) Elevated copper and oxidative stress in Cancer cells as a target for Cancer treatment. Cancer Treat Rev 35(1):32–46PubMed
6.
go back to reference Denoyer D, Masaldan S, La Fontaine S, Cater MA (2015) Targeting copper in Cancer therapy: “copper that cancer”. Metallomics 7(11):1459–1476PubMed Denoyer D, Masaldan S, La Fontaine S, Cater MA (2015) Targeting copper in Cancer therapy: “copper that cancer”. Metallomics 7(11):1459–1476PubMed
7.
go back to reference De Wever O, Derycke L, Hendrix A, De Meerleer G, Godeau F, Depypere H, Bracke M (2007) Soluble Cadherins as cancer biomarkers. Clin Exp Metastasis 24(8):685–697PubMed De Wever O, Derycke L, Hendrix A, De Meerleer G, Godeau F, Depypere H, Bracke M (2007) Soluble Cadherins as cancer biomarkers. Clin Exp Metastasis 24(8):685–697PubMed
8.
go back to reference Tang MKS, Yue PYK, Ip PP, Huang R-L, Lai H-C, Cheung ANY, Tse KY, Ngan HYS, Wong AST (2018) Soluble E-cadherin promotes tumor angiogenesis and localizes to exosome surface. Nat Commun 9(1):1–15 Tang MKS, Yue PYK, Ip PP, Huang R-L, Lai H-C, Cheung ANY, Tse KY, Ngan HYS, Wong AST (2018) Soluble E-cadherin promotes tumor angiogenesis and localizes to exosome surface. Nat Commun 9(1):1–15
9.
go back to reference Thomas C, Mackey MM, Diaz AA, Cox DP (2009) Hydroxyl radical is produced via the Fenton reaction in submitochondrial particles under oxidative stress: implications for diseases associated with Iron accumulation. Redox Rep 14(3):102–108PubMed Thomas C, Mackey MM, Diaz AA, Cox DP (2009) Hydroxyl radical is produced via the Fenton reaction in submitochondrial particles under oxidative stress: implications for diseases associated with Iron accumulation. Redox Rep 14(3):102–108PubMed
10.
go back to reference Fouani L, Menezes SV, Paulson M, Richardson DR, Kovacevic Z (2017) Metals and metastasis: exploiting the role of metals in cancer metastasis to develop novel anti-metastatic agents. Pharmacol Res 115:275–287PubMed Fouani L, Menezes SV, Paulson M, Richardson DR, Kovacevic Z (2017) Metals and metastasis: exploiting the role of metals in cancer metastasis to develop novel anti-metastatic agents. Pharmacol Res 115:275–287PubMed
11.
go back to reference Cichon MA, Radisky DC (2014) ROS-induced epithelial-mesenchymal transition in mammary epithelial cells is mediated by NF-kB-dependent activation of Snail. Oncotarget 5(9):2827–2838PubMedPubMedCentral Cichon MA, Radisky DC (2014) ROS-induced epithelial-mesenchymal transition in mammary epithelial cells is mediated by NF-kB-dependent activation of Snail. Oncotarget 5(9):2827–2838PubMedPubMedCentral
12.
go back to reference Park SY, Yoon SJ, Hakomori S, Jeong YT, Kim JM, Kim JY, Bernert B, Ullman T, Itzkowitz SH, Kim JH, Hakomori SI (2010) N-glycosylation status of beta-haptoglobin in sera of patients with colon cancer, chronic inflammatory disease and normal subjects. Int J Oncol 36(5):1291–1297PubMed Park SY, Yoon SJ, Hakomori S, Jeong YT, Kim JM, Kim JY, Bernert B, Ullman T, Itzkowitz SH, Kim JH, Hakomori SI (2010) N-glycosylation status of beta-haptoglobin in sera of patients with colon cancer, chronic inflammatory disease and normal subjects. Int J Oncol 36(5):1291–1297PubMed
13.
go back to reference Morgan MJ, Liu Z (2011) Crosstalk of reactive oxygen species and NF-κB signalling. Cell Res 21(1):103–115PubMed Morgan MJ, Liu Z (2011) Crosstalk of reactive oxygen species and NF-κB signalling. Cell Res 21(1):103–115PubMed
14.
go back to reference Agrawal A, Romero-Perez D, Jacobsen JA, Villarreal FJ, Cohen SM (2008) Zinc-binding groups modulate selective inhibition of MMPs. Chem Med Chem 3(5):812–820PubMed Agrawal A, Romero-Perez D, Jacobsen JA, Villarreal FJ, Cohen SM (2008) Zinc-binding groups modulate selective inhibition of MMPs. Chem Med Chem 3(5):812–820PubMed
15.
go back to reference Itoh T, Tanioka M, Yoshida H, Yoshioka T, Nishimoto H, Itohara S (1998) Reduced angiogenesis and tumor progression in gelatinase A-deficient mice. Cancer Res 58(5):1048–1051PubMed Itoh T, Tanioka M, Yoshida H, Yoshioka T, Nishimoto H, Itohara S (1998) Reduced angiogenesis and tumor progression in gelatinase A-deficient mice. Cancer Res 58(5):1048–1051PubMed
16.
go back to reference Buss JL, Torti FM, Torti SV (2003) The role of Iron chelation in cancer therapy. Curr Med Chem 10:1021–1034PubMed Buss JL, Torti FM, Torti SV (2003) The role of Iron chelation in cancer therapy. Curr Med Chem 10:1021–1034PubMed
17.
go back to reference Simões RV, Veeraperumal S, Serganova IS, Kruchevsky N, Varshavsky J, Blasberg RG, Ackerstaff E, Koutcher JA (2017) Inhibition of prostate cancer proliferation by Deferiprone. NMR Biomed 30(6):1–11 Simões RV, Veeraperumal S, Serganova IS, Kruchevsky N, Varshavsky J, Blasberg RG, Ackerstaff E, Koutcher JA (2017) Inhibition of prostate cancer proliferation by Deferiprone. NMR Biomed 30(6):1–11
18.
go back to reference Salis O, Bedir A, Kilinc V, Alacam H, Gulten S, Okuyucu A (2014) The anticancer effects of Desferrioxamine on human breast adenocarcinoma and hepatocellular carcinoma cells. Cancer Biomark 14(6):419–426PubMed Salis O, Bedir A, Kilinc V, Alacam H, Gulten S, Okuyucu A (2014) The anticancer effects of Desferrioxamine on human breast adenocarcinoma and hepatocellular carcinoma cells. Cancer Biomark 14(6):419–426PubMed
19.
go back to reference Zhao R, Planalp RP, Ma R, Greene BT, Jones BT, Brechbiel MW, Torti FM, Torti SV (2004) Role of zinc and Iron chelation in apoptosis mediated by Tachpyridine, an anti-cancer Iron chelator. Biochem Pharmacol 67(9):1677–1688PubMed Zhao R, Planalp RP, Ma R, Greene BT, Jones BT, Brechbiel MW, Torti FM, Torti SV (2004) Role of zinc and Iron chelation in apoptosis mediated by Tachpyridine, an anti-cancer Iron chelator. Biochem Pharmacol 67(9):1677–1688PubMed
20.
go back to reference Rejmund M, Mrozek-Wilczkiewicz A, Malarz K, Pyrkosz-Bulska M, Gajcy K, Sajewicz M, Musiol R, Polanski J (2018) Piperazinyl fragment improves anticancer activity of Triapine. PLoS One 13(4):1–25 Rejmund M, Mrozek-Wilczkiewicz A, Malarz K, Pyrkosz-Bulska M, Gajcy K, Sajewicz M, Musiol R, Polanski J (2018) Piperazinyl fragment improves anticancer activity of Triapine. PLoS One 13(4):1–25
21.
go back to reference Richardson DR, Milnes K (1997) The potential of Iron chelators of the pyridoxal Isonicotinoyl Hydrazone class as effective Antiproliferative agents II: the mechanism of action of ligands derived from salicylaldehyde benzoyl hydrazone and 2-Hydroxy-1-Naphthylaldehyde benzoyl Hydrazone. Blood 89(8):3025–3038PubMed Richardson DR, Milnes K (1997) The potential of Iron chelators of the pyridoxal Isonicotinoyl Hydrazone class as effective Antiproliferative agents II: the mechanism of action of ligands derived from salicylaldehyde benzoyl hydrazone and 2-Hydroxy-1-Naphthylaldehyde benzoyl Hydrazone. Blood 89(8):3025–3038PubMed
22.
go back to reference Schwab M (2011) Chelators as anti-Cancer drugs. In: Schwab M (ed) Encyclopedia of cancer. Springer, Berlin, Heidelberg, pp 755–759 Schwab M (2011) Chelators as anti-Cancer drugs. In: Schwab M (ed) Encyclopedia of cancer. Springer, Berlin, Heidelberg, pp 755–759
23.
go back to reference Abeysinghe RD, Roberts PJ, Cooper CE, MacLean KH, Hider RC, Porter JB (1996) The environment of the lipoxygenase Iron binding site explored with novel Hydroxypyridinone Iron chelators. J Biol Chem 271(14):7965–7972PubMed Abeysinghe RD, Roberts PJ, Cooper CE, MacLean KH, Hider RC, Porter JB (1996) The environment of the lipoxygenase Iron binding site explored with novel Hydroxypyridinone Iron chelators. J Biol Chem 271(14):7965–7972PubMed
24.
go back to reference Liu ZD, Lockwood M, Rose S, Theobald AE, Hider RC (2001) Structure-activity investigation of the inhibition of 3-Hydroxypyridin-4-ones on mammalian tyrosine hydroxylase. Biochem Pharmacol 61(3):285–290PubMed Liu ZD, Lockwood M, Rose S, Theobald AE, Hider RC (2001) Structure-activity investigation of the inhibition of 3-Hydroxypyridin-4-ones on mammalian tyrosine hydroxylase. Biochem Pharmacol 61(3):285–290PubMed
25.
go back to reference Tricta F, Uetrecht J, Galanello R, Connelly J, Rozova A, Spino M, Palmblad J (2016) Deferiprone-induced agranulocytosis. Am J Hematol 91(10):1026–1031PubMedPubMedCentral Tricta F, Uetrecht J, Galanello R, Connelly J, Rozova A, Spino M, Palmblad J (2016) Deferiprone-induced agranulocytosis. Am J Hematol 91(10):1026–1031PubMedPubMedCentral
26.
go back to reference Kontoghiorghe CN, Kolnagou A, Kontoghiorghes GJ (2015) Phytochelators intended for clinical use in Iron overload, other diseases of Iron imbalance and free radical pathology. Molecules 20(11):20841–20872PubMedPubMedCentral Kontoghiorghe CN, Kolnagou A, Kontoghiorghes GJ (2015) Phytochelators intended for clinical use in Iron overload, other diseases of Iron imbalance and free radical pathology. Molecules 20(11):20841–20872PubMedPubMedCentral
27.
go back to reference Nguyen BCQ, Tawata S (2016) The chemistry and biological activities of Mimosine: a review. Phytother Res 30:1230–1242PubMed Nguyen BCQ, Tawata S (2016) The chemistry and biological activities of Mimosine: a review. Phytother Res 30:1230–1242PubMed
28.
go back to reference DeWys WD, Hall TC (1973) Anti-tumor effect of the amino acid Mimosine. Eur J Cancer 9(4):281–283PubMed DeWys WD, Hall TC (1973) Anti-tumor effect of the amino acid Mimosine. Eur J Cancer 9(4):281–283PubMed
29.
go back to reference Cabanes J, Garcia-Canovas F, Tudela J, Lozano JA, García-Carmona F (1987) L-mimosine a slow-binding inhibitor of mushroom Tyrosinase. Phytochemistry 26(4):917–919 Cabanes J, Garcia-Canovas F, Tudela J, Lozano JA, García-Carmona F (1987) L-mimosine a slow-binding inhibitor of mushroom Tyrosinase. Phytochemistry 26(4):917–919
30.
go back to reference Soedarjo M, Borthakur D (1996) Simple procedures to remove Mimosine from young leaves, pods and seeds of Leucaena Leucocephala used as food. Int J Food Sci Technol 31(1):97–103 Soedarjo M, Borthakur D (1996) Simple procedures to remove Mimosine from young leaves, pods and seeds of Leucaena Leucocephala used as food. Int J Food Sci Technol 31(1):97–103
31.
go back to reference Vogt G, Böhm R, Segner H (1994) Mimosine-induced cell death and related chromatin changes. J Submicrosc Cytol Pathol 26(3):319–330PubMed Vogt G, Böhm R, Segner H (1994) Mimosine-induced cell death and related chromatin changes. J Submicrosc Cytol Pathol 26(3):319–330PubMed
32.
go back to reference Kulp KS, Vulliet PR (1996) Mimosine blocks cell cycle progression by chelating Iron in asynchronous human breast cancer cells. Toxicol Appl Pharmacol 139(2):356–364PubMed Kulp KS, Vulliet PR (1996) Mimosine blocks cell cycle progression by chelating Iron in asynchronous human breast cancer cells. Toxicol Appl Pharmacol 139(2):356–364PubMed
33.
go back to reference Park SY, Im JS, Park SR, Kim SE, Wang HJ, Lee JK (2012) Mimosine arrests the cell cycle prior to the onset of DNA replication by preventing the binding of human Ctf4/And-1 to chromatin via Hif-1α activation in HeLa cells. Cell Cycle 11(4):761–766PubMed Park SY, Im JS, Park SR, Kim SE, Wang HJ, Lee JK (2012) Mimosine arrests the cell cycle prior to the onset of DNA replication by preventing the binding of human Ctf4/And-1 to chromatin via Hif-1α activation in HeLa cells. Cell Cycle 11(4):761–766PubMed
34.
go back to reference Vogt G, Böhm R, Segner H (1993) Mimosine, a naturally occurring drug interfering primarily with the cell nucleus. J Submicrosc Cytol Pathol 25(2):247–256PubMed Vogt G, Böhm R, Segner H (1993) Mimosine, a naturally occurring drug interfering primarily with the cell nucleus. J Submicrosc Cytol Pathol 25(2):247–256PubMed
35.
go back to reference Gilberts D, Neilson A, Miyazawa H, De Pamphilis M, Burhans W (1995) Mimosine arrests DNA synthesis at replication fork by inhibiting Deoxyribonucleotide metabolism. J Biol Chem 270(16):9597–9606 Gilberts D, Neilson A, Miyazawa H, De Pamphilis M, Burhans W (1995) Mimosine arrests DNA synthesis at replication fork by inhibiting Deoxyribonucleotide metabolism. J Biol Chem 270(16):9597–9606
36.
go back to reference Dai Y, Gold B, Vishwanatha JK, Rhode SL (1994) Mimosine inhibits viral DNA synthesis through ribonucleotide reductase. Virology 205(1):210–216PubMed Dai Y, Gold B, Vishwanatha JK, Rhode SL (1994) Mimosine inhibits viral DNA synthesis through ribonucleotide reductase. Virology 205(1):210–216PubMed
37.
go back to reference Perry C, Sastry R, Nasrallah IM, Stover PJ (2005) Mimosine attenuates serine Hydroxymethyltransferase transcription by chelating zinc: implications for inhibition of DNA replication. J Biol Chem 280(1):396–400PubMed Perry C, Sastry R, Nasrallah IM, Stover PJ (2005) Mimosine attenuates serine Hydroxymethyltransferase transcription by chelating zinc: implications for inhibition of DNA replication. J Biol Chem 280(1):396–400PubMed
38.
go back to reference Lin H, Falchetto R, Mosca PJ, Shabanowitz J, Hunt DF, Hamlin JL (1996) Mimosine targets serine Hydroxymethyltransferase inhibitor of DNA replication in mammalian cells. J Biol Chem 271(5):2548–2556PubMed Lin H, Falchetto R, Mosca PJ, Shabanowitz J, Hunt DF, Hamlin JL (1996) Mimosine targets serine Hydroxymethyltransferase inhibitor of DNA replication in mammalian cells. J Biol Chem 271(5):2548–2556PubMed
39.
go back to reference Hallak M, Dvilansky A, Shpilberg O, Levi I, Mazar J, Nathan I (2004) Mimosine induces apoptosis through metal ion chelation, mitochondrial activation and reactive oxygen species production in human leukemic cells. Blood 104:4481 Hallak M, Dvilansky A, Shpilberg O, Levi I, Mazar J, Nathan I (2004) Mimosine induces apoptosis through metal ion chelation, mitochondrial activation and reactive oxygen species production in human leukemic cells. Blood 104:4481
40.
go back to reference Prabhakaran K, Harris EB, Kirchheimer WF (1973) A specific effect of Mimosine on Melanoma cells. Cytobios 7:245–252PubMed Prabhakaran K, Harris EB, Kirchheimer WF (1973) A specific effect of Mimosine on Melanoma cells. Cytobios 7:245–252PubMed
41.
go back to reference Khwaja TA, Hall TC, Sheikh KMA (1976) Antitumor activity of Mimosine and Mimosine hydrochloride against B16 melanoma in BDF mice. In: Hellmann K, Connors TA (eds) Chemotherapy, vol 8. Springer, Boston Khwaja TA, Hall TC, Sheikh KMA (1976) Antitumor activity of Mimosine and Mimosine hydrochloride against B16 melanoma in BDF mice. In: Hellmann K, Connors TA (eds) Chemotherapy, vol 8. Springer, Boston
42.
go back to reference Prabhakaran K, Harris EB, Kircheimer WF (1969) Suppression of melanoma development and inhibition of phenoloxidase by mimosine. Cytobios 1A:3–5 Prabhakaran K, Harris EB, Kircheimer WF (1969) Suppression of melanoma development and inhibition of phenoloxidase by mimosine. Cytobios 1A:3–5
43.
go back to reference Fraga S, Serrão MP, Soares-da-Silva P (2002) L-type amino acid transporters in two intestinal epithelial cell lines function as exchangers with neutral amino acids. J Nutr 132(4):733–738PubMed Fraga S, Serrão MP, Soares-da-Silva P (2002) L-type amino acid transporters in two intestinal epithelial cell lines function as exchangers with neutral amino acids. J Nutr 132(4):733–738PubMed
44.
go back to reference Augustyn E, Finke K, Zur AA, Hansen L, Heeren N, Chien HC, Lin L, Giacomini KM, Colas C, Schlessinger A, Thomas A (2016) LAT-1 activity of meta substituted phenylalanine and tyrosine analogs. Bioorg Med Chem Lett 26:2616–2621PubMedPubMedCentral Augustyn E, Finke K, Zur AA, Hansen L, Heeren N, Chien HC, Lin L, Giacomini KM, Colas C, Schlessinger A, Thomas A (2016) LAT-1 activity of meta substituted phenylalanine and tyrosine analogs. Bioorg Med Chem Lett 26:2616–2621PubMedPubMedCentral
45.
go back to reference Chien HC, Colas C, Finke K, Springer S, Stoner L, Zur AA, Venteicher B, Campbell J, Hall C, Flint A, Augustyn E, Hernandez C, Heeren N, Hansen L, Anthony A, Bauer J, Fotiadis D, Schlessinger A, Giacomini KM, Thomas AA (2018) Reevaluating the substrate specificity of the L-type amino acid transporter (LAT1). J Med Chem 61:7358–7373PubMedPubMedCentral Chien HC, Colas C, Finke K, Springer S, Stoner L, Zur AA, Venteicher B, Campbell J, Hall C, Flint A, Augustyn E, Hernandez C, Heeren N, Hansen L, Anthony A, Bauer J, Fotiadis D, Schlessinger A, Giacomini KM, Thomas AA (2018) Reevaluating the substrate specificity of the L-type amino acid transporter (LAT1). J Med Chem 61:7358–7373PubMedPubMedCentral
46.
go back to reference Geier EG, Schlessinger A, Fan H, Gable JE, Irwin JJ, Sali A, Giacomini KM (2013) Structure-based ligand discovery for the large-neutral amino acid transporter 1 LAT-1. Proc Natl Acad Sci U S A 110:5480–5485PubMedPubMedCentral Geier EG, Schlessinger A, Fan H, Gable JE, Irwin JJ, Sali A, Giacomini KM (2013) Structure-based ligand discovery for the large-neutral amino acid transporter 1 LAT-1. Proc Natl Acad Sci U S A 110:5480–5485PubMedPubMedCentral
47.
go back to reference Lin J, Raoof DA, Thomas DG, Greenson JK, Giordano TJ, Robinson GS, Bourner MJ, Bauer CT, Orringer MB, Beer DG (2004) L-type amino acid Transporter-1 overexpression and Melphalan sensitivity in Barrett’s adenocarcinoma. Neoplasia 6(1):74–84PubMedPubMedCentral Lin J, Raoof DA, Thomas DG, Greenson JK, Giordano TJ, Robinson GS, Bourner MJ, Bauer CT, Orringer MB, Beer DG (2004) L-type amino acid Transporter-1 overexpression and Melphalan sensitivity in Barrett’s adenocarcinoma. Neoplasia 6(1):74–84PubMedPubMedCentral
49.
go back to reference Fulda S (2015) Targeting extrinsic apoptosis in cancer: challenges and opportunities. Semin Cell Dev Biol 39:20–25PubMed Fulda S (2015) Targeting extrinsic apoptosis in cancer: challenges and opportunities. Semin Cell Dev Biol 39:20–25PubMed
50.
go back to reference Kantari C, Walczak H (2011) Caspase-8 and bid: caught in the act between death receptors and mitochondria. Biochim Biophys Acta 1813(4):558–563PubMed Kantari C, Walczak H (2011) Caspase-8 and bid: caught in the act between death receptors and mitochondria. Biochim Biophys Acta 1813(4):558–563PubMed
51.
go back to reference Li P, Zhou L, Zhao T, Liu X, Zhang P, Liu Y, Zheng X, Li Q (2017) Caspase-9: structure, mechanism and clinical application. Oncotarget 8(14):23996–24008PubMedPubMedCentral Li P, Zhou L, Zhao T, Liu X, Zhang P, Liu Y, Zheng X, Li Q (2017) Caspase-9: structure, mechanism and clinical application. Oncotarget 8(14):23996–24008PubMedPubMedCentral
52.
go back to reference Shakeri R, Kheirollahi A, Davoodi J (2017) Apaf-1: regulation and function in cell death. Biochimie 135:111–125PubMed Shakeri R, Kheirollahi A, Davoodi J (2017) Apaf-1: regulation and function in cell death. Biochimie 135:111–125PubMed
54.
go back to reference Zhou M, Li Y, Hu Q, Bai X, Huang W, Yan C, Scheres SH, Shi Y (2015) Atomic structure of the apoptosome: mechanism of cytochrome c- and dATP mediated activation of Apaf-1. Genes Dev 29(22):2349–2361PubMedPubMedCentral Zhou M, Li Y, Hu Q, Bai X, Huang W, Yan C, Scheres SH, Shi Y (2015) Atomic structure of the apoptosome: mechanism of cytochrome c- and dATP mediated activation of Apaf-1. Genes Dev 29(22):2349–2361PubMedPubMedCentral
55.
go back to reference Hu Q, Chen W, Zhen Y, Shi Y (2013) Proteolytic processing of the caspase-9 zymogen is required for Apoptosome-mediated activation of caspase. J Biol Chem 288(21):15142–15147PubMedPubMedCentral Hu Q, Chen W, Zhen Y, Shi Y (2013) Proteolytic processing of the caspase-9 zymogen is required for Apoptosome-mediated activation of caspase. J Biol Chem 288(21):15142–15147PubMedPubMedCentral
56.
go back to reference Würstle ML, Rehm M (2014) A systems biology analysis of apoptosome formation and apoptosis execution supports allosteric procaspase-9 activation. J Biol Chem 289(38):26277–26289PubMedPubMedCentral Würstle ML, Rehm M (2014) A systems biology analysis of apoptosome formation and apoptosis execution supports allosteric procaspase-9 activation. J Biol Chem 289(38):26277–26289PubMedPubMedCentral
57.
go back to reference Yuan S, Yu X, Asaram JM, Heuser JE, Ludtke SL, Akey CW (2011) The holo-apoptosome: activation of procaspase-9 and interactions with caspase-3. Structure 19:1084–1096PubMedPubMedCentral Yuan S, Yu X, Asaram JM, Heuser JE, Ludtke SL, Akey CW (2011) The holo-apoptosome: activation of procaspase-9 and interactions with caspase-3. Structure 19:1084–1096PubMedPubMedCentral
58.
go back to reference Wright KM, Deshmukh M, Vaughn AE (2007) Apoptosome dependent caspase-3 activation pathway is non-redundant and necessary for apoptosis in sympathic neurons. Cell Death Differ 14:625–633PubMed Wright KM, Deshmukh M, Vaughn AE (2007) Apoptosome dependent caspase-3 activation pathway is non-redundant and necessary for apoptosis in sympathic neurons. Cell Death Differ 14:625–633PubMed
59.
go back to reference Zou H, Yang R, Hao J, Wang J, Sun C, Fesik SW, Wu JC, Tomaselli KJ, Armstrong RCJ (2003) Regulation of the Apaf-1/Caspase-9 apoptosome by Caspase-3 and XIAP. J Biol Chem 278(10):8091–8098PubMed Zou H, Yang R, Hao J, Wang J, Sun C, Fesik SW, Wu JC, Tomaselli KJ, Armstrong RCJ (2003) Regulation of the Apaf-1/Caspase-9 apoptosome by Caspase-3 and XIAP. J Biol Chem 278(10):8091–8098PubMed
60.
go back to reference Kang YJ, Kuo C-F, Majd S (2017) Nanoparticle-based deliver of an anti-proliferative metal chelator to tumor cells. IEEE:309–312 Kang YJ, Kuo C-F, Majd S (2017) Nanoparticle-based deliver of an anti-proliferative metal chelator to tumor cells. IEEE:309–312
61.
go back to reference Tsuma-Kaneko M, Sawanobori M, Kawakami S, Uno T, Nakamura Y, Onizuka M, Ando K, Kawada H (2018) Iron removal enhances vitamin C-induced apoptosis and growth inhibition of K-562 leukemic cells. Sci Rep 8:17377PubMedPubMedCentral Tsuma-Kaneko M, Sawanobori M, Kawakami S, Uno T, Nakamura Y, Onizuka M, Ando K, Kawada H (2018) Iron removal enhances vitamin C-induced apoptosis and growth inhibition of K-562 leukemic cells. Sci Rep 8:17377PubMedPubMedCentral
62.
go back to reference Xu LL, Hu PP, Kong X, Hider RC, Zhou T, Dai ZY (2014) 3-Hydroxypyridinone-l-phenylanaline conjugates with antimicrobial and tyrosinase activities as potential shrimp preservatives. Int J Food Sci Technol 49(3):797–803 Xu LL, Hu PP, Kong X, Hider RC, Zhou T, Dai ZY (2014) 3-Hydroxypyridinone-l-phenylanaline conjugates with antimicrobial and tyrosinase activities as potential shrimp preservatives. Int J Food Sci Technol 49(3):797–803
63.
64.
go back to reference Slominski A, Zmijewski MA, Pawelek J (2012) L-tyrosine and L-dihydroxyphenylalanine as hormone-like regulators of melanocyte fuctions. Pigment Cell Melanoma Res 25:14–27PubMed Slominski A, Zmijewski MA, Pawelek J (2012) L-tyrosine and L-dihydroxyphenylalanine as hormone-like regulators of melanocyte fuctions. Pigment Cell Melanoma Res 25:14–27PubMed
65.
go back to reference Brozyna AA, Jozwicki W, Roszkowski K, Filipiak J, Slominski AT (2016) Melanine content in melanoma metastases affects the outcome radiotherapy. Oncotarget 7:17844–17853PubMedPubMedCentral Brozyna AA, Jozwicki W, Roszkowski K, Filipiak J, Slominski AT (2016) Melanine content in melanoma metastases affects the outcome radiotherapy. Oncotarget 7:17844–17853PubMedPubMedCentral
66.
go back to reference Prota G, D’Jschia M, Mascagna D (1994) Melanogenesis as a targeting strategy against metastatic melanoma: a reassessment. Melanoma Res 4:351–358PubMed Prota G, D’Jschia M, Mascagna D (1994) Melanogenesis as a targeting strategy against metastatic melanoma: a reassessment. Melanoma Res 4:351–358PubMed
67.
go back to reference Novellino L, Napolitano A, Prota G (1999) 5,6-Dihydroxyindoles in the Fenton chemistry: a model study of melanin precursors in oxidative stress and hyperpigmentary processes. Chem Res Toxicol 12:985–992PubMed Novellino L, Napolitano A, Prota G (1999) 5,6-Dihydroxyindoles in the Fenton chemistry: a model study of melanin precursors in oxidative stress and hyperpigmentary processes. Chem Res Toxicol 12:985–992PubMed
68.
go back to reference Inbaraj J, Gandhidasan R, Murugesan R (1999) Cytotoxicity and superoxide anion generation by some naturally occurring quinones. Free Radic Biol Med 26:1072–1078 Inbaraj J, Gandhidasan R, Murugesan R (1999) Cytotoxicity and superoxide anion generation by some naturally occurring quinones. Free Radic Biol Med 26:1072–1078
69.
go back to reference Zhou P, Qin J, Li Y, Li G, Wang Y, Zhan N, Chen P, Li C (2017) Combination therapy of PKCζ and COX-2 inhibitors synergistically suppress melanoma metastasis. JECCR 36(115):1–12 Zhou P, Qin J, Li Y, Li G, Wang Y, Zhan N, Chen P, Li C (2017) Combination therapy of PKCζ and COX-2 inhibitors synergistically suppress melanoma metastasis. JECCR 36(115):1–12
70.
go back to reference Meyskens FL, McNulty SE, Buckmeier JA, Tohidian NB, Spillane TJ, KAhlon RS, Gonzales RI (2001) Aberrant redox regulation in human malignant metastatic melanoma cells compare to normal melanocytes. Free Radic Biol Med 31(6):799–808PubMed Meyskens FL, McNulty SE, Buckmeier JA, Tohidian NB, Spillane TJ, KAhlon RS, Gonzales RI (2001) Aberrant redox regulation in human malignant metastatic melanoma cells compare to normal melanocytes. Free Radic Biol Med 31(6):799–808PubMed
71.
go back to reference Mohammadpour M, Sadeghi A, Fassihi A, Saghaie L (2012) Synthesis and antioxidant evaluation of some novel ortho-hydroxypyridine-4-one iron chelators. Res Pharm Sci 7(3):171–179PubMedPubMedCentral Mohammadpour M, Sadeghi A, Fassihi A, Saghaie L (2012) Synthesis and antioxidant evaluation of some novel ortho-hydroxypyridine-4-one iron chelators. Res Pharm Sci 7(3):171–179PubMedPubMedCentral
72.
go back to reference Lu N, Chen W, PEng YY (2011) Effects of glutathione, Trolox and desferrioxamine on hemoglobin induced protein oxidative damage: anti-oxidant or pro-oxidant? Eur J Pharmacol 659(2–3):95–101PubMed Lu N, Chen W, PEng YY (2011) Effects of glutathione, Trolox and desferrioxamine on hemoglobin induced protein oxidative damage: anti-oxidant or pro-oxidant? Eur J Pharmacol 659(2–3):95–101PubMed
73.
go back to reference Qiao S, Murakami K, Zhao Q, Wank B, Seo H, Yamashita H, Li X, Iwamoto T, Ichiharam M, Yoshino M (2012) Mimosine-induced apoptosis in C6 glioma cells requires the release of mitochondria-derived reactive oxygen species and p38. JNK activation. Neurochem Res 37:412–427 Qiao S, Murakami K, Zhao Q, Wank B, Seo H, Yamashita H, Li X, Iwamoto T, Ichiharam M, Yoshino M (2012) Mimosine-induced apoptosis in C6 glioma cells requires the release of mitochondria-derived reactive oxygen species and p38. JNK activation. Neurochem Res 37:412–427
74.
go back to reference Akladios FN, Andrew SD, Parkinson CJ (2016) Investigation of the cytotoxic implications of metal chelators against melanoma and approaches to improve the cytotoxic profiles of metal coordinating agents. Biometals 29(5):789–805PubMed Akladios FN, Andrew SD, Parkinson CJ (2016) Investigation of the cytotoxic implications of metal chelators against melanoma and approaches to improve the cytotoxic profiles of metal coordinating agents. Biometals 29(5):789–805PubMed
75.
go back to reference Xu Y, Cai L (2018) L-mimosine induces caspase-9-mediated apoptosis in human osteosarcoma cells. Mol Med Rep 17:4695–4701PubMed Xu Y, Cai L (2018) L-mimosine induces caspase-9-mediated apoptosis in human osteosarcoma cells. Mol Med Rep 17:4695–4701PubMed
76.
go back to reference Zalatnai A, Bocsi J (2003) Mimosine, a plant-derived amino acid induces apoptosis in human pancreatic cancer xenografts. Anticancer Res 23:4007–4009PubMed Zalatnai A, Bocsi J (2003) Mimosine, a plant-derived amino acid induces apoptosis in human pancreatic cancer xenografts. Anticancer Res 23:4007–4009PubMed
77.
go back to reference Meyskens FL, McNulty SE, Buckmeier JA, Tohidian NB, Spillane TJ, KAhlon RS, Gonzales RI (2001) Aberrant redox regulation in human malignant metastatic melanoma cells compared to normal melanocytes. Free Radic Biol Med 31(6):799–808PubMed Meyskens FL, McNulty SE, Buckmeier JA, Tohidian NB, Spillane TJ, KAhlon RS, Gonzales RI (2001) Aberrant redox regulation in human malignant metastatic melanoma cells compared to normal melanocytes. Free Radic Biol Med 31(6):799–808PubMed
78.
go back to reference Sauer H, Wartenberg M, Hescheier J (2001) Reactive oxygen species as intracellular messengers during cell growth and differentiation. Cell Physiol Biochem 11(4):173–186PubMed Sauer H, Wartenberg M, Hescheier J (2001) Reactive oxygen species as intracellular messengers during cell growth and differentiation. Cell Physiol Biochem 11(4):173–186PubMed
79.
go back to reference Redza-Dutordoir M, Averill-Bates DA (2016) Activation of apoptosis signalling pathways by reactive oxygen species. Biochim Biophys Acta 1863(12):2977–2992PubMed Redza-Dutordoir M, Averill-Bates DA (2016) Activation of apoptosis signalling pathways by reactive oxygen species. Biochim Biophys Acta 1863(12):2977–2992PubMed
80.
go back to reference Simonart T, Degraef C, Andrei G, Mosselmans R (2000) Iron chelators inhibit the growth and induce the apoptosis of Kaposi’s sarcoma cells and of their putative endothelial precursors. J Invest Dermatol 115(5):893–900PubMed Simonart T, Degraef C, Andrei G, Mosselmans R (2000) Iron chelators inhibit the growth and induce the apoptosis of Kaposi’s sarcoma cells and of their putative endothelial precursors. J Invest Dermatol 115(5):893–900PubMed
81.
go back to reference Chang HC, Lee TH, Chuang LY, Yen MH, Hung WC (1999) Inhibitory effect of mimosine on proliferation of human lung cancer cells is mediated by multiple mechanisms. Cancer Lett 145:1–8PubMed Chang HC, Lee TH, Chuang LY, Yen MH, Hung WC (1999) Inhibitory effect of mimosine on proliferation of human lung cancer cells is mediated by multiple mechanisms. Cancer Lett 145:1–8PubMed
Metadata
Title
Anticancer activity of a novel methylated analogue of L-mimosine against an in vitro model of human malignant melanoma
Authors
Sotiris Kyriakou
Melina Mitsiogianni
Theodora Mantso
William Cheung
Stephen Todryk
Stephany Veuger
Aglaia Pappa
David Tetard
Mihalis I. Panayiotidis
Publication date
01-06-2020
Publisher
Springer US
Published in
Investigational New Drugs / Issue 3/2020
Print ISSN: 0167-6997
Electronic ISSN: 1573-0646
DOI
https://doi.org/10.1007/s10637-019-00809-0

Other articles of this Issue 3/2020

Investigational New Drugs 3/2020 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine