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

A review of ceramide analogs as potential anticancer agents

    Jiawang Liu

    Department of Chemistry, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, LA 70125, USA

    ,
    Barbara S Beckman

    Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA

    &
    Maryam Foroozesh

    * Author for correspondence

    Department of Chemistry, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, LA 70125, USA.

    Published Online:https://doi.org/10.4155/fmc.13.107

    Ceramide serves as a central mediator in sphingolipid metabolism and signaling pathways, regulating many fundamental cellular responses. It is referred to as a ‘tumor suppressor lipid’, since it powerfully potentiates signaling events that drive apoptosis, cell cycle arrest, and autophagic responses. In the typical cancer cell, ceramide levels and signaling are usually suppressed by overexpression of ceramide-metabolizing enzymes or downregulation of ceramide-generating enzymes. However, chemotherapeutic drugs as well as radiotherapy increase intracellular ceramide levels, while exogenously treating cancer cells with short-chain ceramides leads to anticancer effects. All evidence currently points to the fact that the upregulation of ceramide levels is a promising anticancer strategy. In this review, we exhibit many anticancer ceramide analogs as downstream receptor agonists and ceramide-metabolizing enzyme inhibitors.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    References

    • Huwiler A, Zangemeister-Wittke U. Targeting the conversion of ceramide to sphingosine 1-phosphate as a novel strategy for cancer therapy. Crit. Rev. Oncol. Hematol.63(2),150–159 (2007).
    • Takabe K, Paugh SW, Milstien S, Spiegel S. ‘Inside-out’ signaling of sphingosine-1-phosphate: therapeutic targets. Pharmacol. Rev.60(2),181–195 (2008).
    • Huwiler A, Kolter T, Pfeilschifter J, Sandhoff K. Physiology and pathophysiology of sphingolipid metabolism and signaling. Biochim. Biophys. Acta1485(2–3),63–99 (2000).
    • Ogretmen B, Hannun YA. Biologically active sphingolipids in cancer pathogenesis and treatment. Nat. Rev. Cancer4(8),604–616 (2004).
    • Zhou H, Summers SA, Birnbaum MJ, Pittman RN. Inhibition of AKT kinase by cell-permeable ceramide and its implications for ceramide-induced apoptosis. J. Biol. Chem.273(26),16568–16575 (1998).
    • Bourbon NA, Sandirasegarane L, Kester M. Ceramide-induced inhibition of AKT is mediated through protein kinase Czeta: implications for growth arrest. J. Biol. Chem.277(5),3286–3292 (2002).
    • Wang G, Silva J, Krishnamurthy K, Tran E, Condie BG, Bieberich E. Direct binding to ceramide activates protein kinase Czeta before the formation of a pro-apoptotic complex with PAR-4 in differentiating stem cells. J. Biol. Chem.280(28),26415–26424 (2005).
    • Deng X, Gao F, May WS. Protein phosphatase 2A inactivates Bcl2’s antiapoptotic function by dephosphorylation and up-regulation of Bcl2-p53 binding. Blood113(2),422–428 (2009).
    • Heinrich M, Neumeyer J, Jakob M et al. Cathepsin D links TNF-induced acid sphingomyelinase to Bid-mediated caspase-9 and -3 activation. Cell Death Differ.11(5),550–563 (2004).
    • 10  Lee H, Rotolo JA, Mesicek J et al. Mitochondrial ceramide-rich macrodomains functionalize Bax upon irradiation. PLoS ONE6(6),e19783 (2011).
    • 11  Von Haefen C, Wieder T, Gillissen B et al. Ceramide induces mitochondrial activation and apoptosis via a Bax-dependent pathway in human carcinoma cells. Oncogene21(25),4009–4019 (2002).
    • 12  Beckham TH, Lu P, Jones EE et al. LCL124, a cationic analog of ceramide, selectively induces pancreatic cancer cell death by accumulating in mitochondria. J. Pharmacol. Exp. Ther.344(1),167–178 (2013).▪ Positively charged ceramide analog LCL124.
    • 13  Dahm F, Bielawska A, Nocito A et al. Mitochondrially targeted ceramide LCL-30 inhibits colorectal cancer in mice. Br. J. Cancer98(1),98–105 (2008).
    • 14  Chalfant CE, Szulc Z, Roddy P, Bielawska A, Hannun YA. The structural requirements for ceramide activation of serine-threonine protein phosphatases. J. Lipid Res.45(3),496–506 (2004).
    • 15  Bielawska A, Greenberg MS, Perry D et al. (1S,2R)-D-erythro-2-(N-myristoylamino)-1-phenyl-1-propanol as an inhibitor of ceramidase. J. Biol. Chem.271(21),12646–12654 (1996).
    • 16  Azuma H, Ijichi S, Kataoka M et al. Short-chain 3-ketoceramides, strong apoptosis inducers against human leukemia HL-60 cells. Bioorg. Med. Chem.15(8),2860–2867 (2007).
    • 17  Bernardo K, Hurwitz R, Zenk T et al. Purification, characterization, and biosynthesis of human acid ceramidase. J. Biol. Chem.270(19),11098–11102 (1995).
    • 18  El Bawab S, Roddy P, Qian T, Bielawska A, Lemasters JJ, Hannun YA. Molecular cloning and characterization of a human mitochondrial ceramidase. J. Biol. Chem.275(28),21508–21513 (2000).
    • 19  Tani M, Okino N, Mitsutake S, Tanigawa T, Izu H, Ito M. Purification and characterization of a neutral ceramidase from mouse liver. A single protein catalyzes the reversible reaction in which ceramide is both hydrolyzed and synthesized. J. Biol. Chem.275(5),3462–3468 (2000).
    • 20  Okino N, Tani M, Imayama S, Ito M. Purification and characterization of a novel ceramidase from Pseudomonas aeruginosa.?. J. Biol. Chem.273(23),14368–14373 (1998).
    • 21  Raisova M, Goltz G, Bektas M et al. Bcl-2 overexpression prevents apoptosis induced by ceramidase inhibitors in malignant melanoma and HaCaT keratinocytes. FEBS Lett.516(1–3),47–52 (2002).
    • 22  Samsel L, Zaidel G, Drumgoole HM et al. The ceramide analog, B13, induces apoptosis in prostate cancer cell lines and inhibits tumor growth in prostate cancer xenografts. Prostate58(4),382–393 (2004).
    • 23  Granot T, Milhas D, Carpentier S et al. Caspase-dependent and -independent cell death of Jurkat human leukemia cells induced by novel synthetic ceramide analogs. Leukemia20(3),392–399 (2006).
    • 24  Ichikawa S, Sakiyama H, Suzuki G, Hidari KI, Hirabayashi Y. Expression cloning of a cDNA for human ceramide glucosyltransferase that catalyzes the first glycosylation step of glycosphingolipid synthesis. Proc. Natl Acad. Sci. USA93(10),4638–4643 (1996).
    • 25  Song M, Zang W, Zhang B, Cao J, Yang G. GCS overexpression is associated with multidrug resistance of human HCT-8 colon cancer cells. J. Exp. Clin. Cancer Res. CR31,23 (2012).
    • 26  Liu YY, Han TY, Giuliano AE, Cabot MC. Expression of glucosylceramide synthase, converting ceramide to glucosylceramide, confers adriamycin resistance in human breast cancer cells. J. Biol. Chem.274(2),1140–1146 (1999).
    • 27  Liu YY, Yu JY, Yin D et al. A role for ceramide in driving cancer cell resistance to doxorubicin. FASEB J.22(7),2541–2551 (2008).
    • 28  Liu YY, Gupta V, Patwardhan GA et al. Glucosylceramide synthase upregulates MDR1 expression in the regulation of cancer drug resistance through cSrc and beta-catenin signaling. Mol. Cancer9,145 (2010).
    • 29  Gouaze-Andersson V, Yu JY, Kreitenberg AJ, Bielawska A, Giuliano AE, Cabot MC. Ceramide and glucosylceramide upregulate expression of the multidrug resistance gene MDR1 in cancer cells. Biochim. Biophys. Acta1771(12),1407–1417 (2007).
    • 30  Gouazé V, Liu YY, Prickett CS, Yu JY, Giuliano AE, Cabot MC. Glucosylceramide synthase blockade down-regulates P-glycoprotein and resensitizes multidrug-resistant breast cancer cells to anticancer drugs. Cancer Res.65(9),3861–3867 (2005).
    • 31  Sietsma H, Veldman RJ, Kolk D et al. 1-phenyl-2-decanoylamino-3-morpholino-1-propanol chemosensitizes neuroblastoma cells for taxol and vincristine. Clin. Cancer Res.6(3),942–948 (2000).
    • 32  Dijkhuis AJ, Klappe K, Jacobs S et al. PDMP sensitizes neuroblastoma to paclitaxel by inducing aberrant cell cycle progression leading to hyperploidy. Mol. Cancer Ther.5(3),593–601 (2006).
    • 33  Granado MH, Gangoiti P, Ouro A, Arana L, Gomez-Munoz A. Ceramide 1-phosphate inhibits serine palmitoyltransferase and blocks apoptosis in alveolar macrophages. Biochim. Biophys. Acta1791(4),263–272 (2009).
    • 34  Gomez-Munoz A, Kong JY, Salh B, Steinbrecher UP. Ceramide-1-phosphate blocks apoptosis through inhibition of acid sphingomyelinase in macrophages. J. Lipid Res.45(1),99–105 (2004).
    • 35  Gangoiti P, Granado MH, Wang SW, Kong JY, Steinbrecher UP, Gomez-Munoz A. Ceramide 1-phosphate stimulates macrophage proliferation through activation of the PI3-kinase/PKB, JNK and ERK1/2 pathways. Cell. Signal.20(4),726–736 (2008).
    • 36  Gangoiti P, Bernacchioni C, Donati C et al. Ceramide 1-phosphate stimulates proliferation of C2C12 myoblasts. Biochimie94(3),597–607 (2012).
    • 37  Sauer B, Gonska H, Manggau M et al. Sphingosine 1-phosphate is involved in cytoprotective actions of calcitriol in human fibroblasts and enhances the intracellular Bcl-2/Bax rheostat. Pharmazie60(4),298–304 (2005).
    • 38  Avery K, Avery S, Shepherd J, Heath PR, Moore H. Sphingosine-1-phosphate mediates transcriptional regulation of key targets associated with survival, proliferation, and pluripotency in human embryonic stem cells. Stem Cells Dev.17(6),1195–1205 (2008).
    • 39  Jürgensmeier JM, Xie Z, Deveraux Q, Ellerby L, Bredesen D, Reed JC. Bax directly induces release of cytochrome c from isolated mitochondria. Proc. Natl Acad. Sci. USA95(9),4997–5002 (1998).
    • 40  Bektas M, Jolly PS, Müller C, Eberle J, Spiegel S, Geilen CC. Sphingosine kinase activity counteracts ceramide-mediated cell death in human melanoma cells: role of Bcl-2 expression. Oncogene24(1),178–187 (2005).
    • 41  Pyne NJ, Pyne S. Sphingosine 1-phosphate and cancer. Nat. Rev. Cancer10(7),489–503 (2010).
    • 42  Mitra P, Oskeritzian CA, Payne SG, Beaven MA, Milstien S, Spiegel S. Role of ABCC1 in export of sphingosine-1-phosphate from mast cells. Proc. Natl Acad. Sci. USA103(44),16394–16399 (2006).
    • 43  Calise S, Blescia S, Cencetti F, Bernacchioni C, Donati C, Bruni P. Sphingosine 1-phosphate stimulates proliferation and migration of satellite cells: role of S1P receptors. Biochim. Biophys. Acta1823(2),439–450 (2012).
    • 44  Wang F, Van Brocklyn JR, Hobson JP et al. Sphingosine 1-phosphate stimulates cell migration through a G(i)-coupled cell surface receptor. Potential involvement in angiogenesis. J. Biol. Chem.274(50),35343–35350 (1999).
    • 45  Sugimoto N, Takuwa N, Okamoto H, Sakurada S, Takuwa Y. Inhibitory and stimulatory regulation of Rac and cell motility by the G12/13-Rho and Gi pathways integrated downstream of a single G protein-coupled sphingosine-1-phosphate receptor isoform. Mol. Cell. Biol.23(5),1534–1545 (2003).
    • 46  Hsu A, Zhang W, Lee JF et al. Sphingosine-1-phosphate receptor-3 signaling up-regulates epidermal growth factor receptor and enhances epidermal growth factor receptor-mediated carcinogenic activities in cultured lung adenocarcinoma cells. Int. J. Oncol.40(5),1619–1626 (2012).
    • 47  Kim ES, Kim JS, Kim SG, Hwang S, Lee CH, Moon A. Sphingosine 1-phosphate regulates matrix metalloproteinase-9 expression and breast cell invasion through S1P3-Gαq coupling. J. Cell. Sci.124(Pt 13),2220–2230 (2011).
    • 48  English D, Welch Z, Kovala AT et al. Sphingosine 1-phosphate released from platelets during clotting accounts for the potent endothelial cell chemotactic activity of blood serum and provides a novel link between hemostasis and angiogenesis. FASEB J.14(14),2255–2265 (2000).
    • 49  Bieberich E, Hu B, Silva J et al. Synthesis and characterization of novel ceramide analogs for induction of apoptosis in human cancer cells. Cancer Lett.181(1),55–64 (2002).
    • 50  Nicholson KM, Quinn DM, Kellett GL, Warr JR. Preferential killing of multidrug-resistant KB cells by inhibitors of glucosylceramide synthase. Br. J. Cancer81(3),423–430 (1999).
    • 51  Wang JD, Takahara S, Nonomura N et al. Early induction of apoptosis in androgen-independent prostate cancer cell line by FTY720 requires caspase-3 activation. Prostate40(1),50–55 (1999).
    • 52  Stover T, Kester M. Liposomal delivery enhances short-chain ceramide-induced apoptosis of breast cancer cells. J. Pharmacol. Exp. Ther.307(2),468–475 (2003).
    • 53  Stover TC, Sharma A, Robertson GP, Kester M. Systemic delivery of liposomal short-chain ceramide limits solid tumor growth in murine models of breast adenocarcinoma. Clin. Cancer Res.11(9),3465–3474 (2005).
    • 54  Zolnik BS, Stern ST, Kaiser JM et al. Rapid distribution of liposomal short-chain ceramide in vitro and in vivo.?. Drug Metab. Dispos.36(8),1709–1715 (2008).
    • 55  Morad SA, Levin JC, Shanmugavelandy SS et al. Ceramide – antiestrogen nanoliposomal combinations – novel impact of hormonal therapy in hormone-insensitive breast cancer. Mol. Cancer Ther.11(11),2352–2361 (2012).
    • 56  Bielawska A, Crane HM, Liotta D, Obeid LM, Hannun YA. Selectivity of ceramide-mediated biology. Lack of activity of erythro-dihydroceramide. J. Biol. Chem.268(35),26226–26232 (1993).▪▪ Stereospecific activity of ceramide analogs.
    • 57  Bielawska A, Linardic CM, Hannun YA. Ceramide-mediated biology. Determination of structural and stereospecific requirements through the use of N-acyl-phenylaminoalcohol analogs. J. Biol. Chem.267(26),18493–18497 (1992).
    • 58  Karasavvas N, Erukulla RK, Bittman R, Lockshin R, Zakeri Z. Stereospecific induction of apoptosis in U937 cells by N-octanoyl-sphingosine stereoisomers and N-octyl-sphingosine. The ceramide amide group is not required for apoptosis. Eur. J. Biochem.236(2),729–737 (1996).
    • 59  Szulc ZM, Bai A, Bielawski J et al. Synthesis, NMR characterization and divergent biological actions of 2´-hydroxy-ceramide/dihydroceramide stereoisomers in MCF7 cells. Bioorg. Med. Chem.18(21),7565–7579 (2010).
    • 60  Lim KG, Sun C, Bittman R, Pyne NJ, Pyne S. (R)-FTY720 methyl ether is a specific sphingosine kinase 2 inhibitor: effect on sphingosine kinase 2 expression in HEK 293 cells and actin rearrangement and survival of MCF-7 breast cancer cells. Cell. Signal.23(10),1590–1595 (2011).
    • 61  Buehrer BM, Bell RM. Inhibition of sphingosine kinase in vitro and in platelets. Implications for signal transduction pathways. J. Biol. Chem.267(5),3154–3159 (1992).
    • 62  Usta J, El Bawab S, Roddy P et al. Structural requirements of ceramide and sphingosine based inhibitors of mitochondrial ceramidase. Biochemistry40(32),9657–9668 (2001).
    • 63  Vunnam RR, Radin NS. Analogs of ceramide that inhibit glucocerebroside synthetase in mouse brain. Chem. Phys. Lipids26(3),265–278 (1980).▪▪ Discovery of RV-583 as a competitive glucosylceramide synthase inhibitor.
    • 64  Selzner M, Bielawska A, Morse MA et al. Induction of apoptotic cell death and prevention of tumor growth by ceramide analogues in metastatic human colon cancer. Cancer Res.61(3),1233–1240 (2001).
    • 65  Dagan A, Wang C, Fibach E, Gatt S. Synthetic, non-natural sphingolipid analogs inhibit the biosynthesis of cellular sphingolipids, elevate ceramide and induce apoptotic cell death. Biochim. Biophys. Acta1633(3),161–169 (2003).
    • 66  Holman DH, Turner LS, El-Zawahry A et al. Lysosomotropic acid ceramidase inhibitor induces apoptosis in prostate cancer cells. Cancer Chemother. Pharmacol.61(2),231–242 (2008).
    • 67  Bhabak KP, Kleuser B, Huwiler A, Arenz C. Effective inhibition of acid and neutral ceramidases by novel B-13 and LCL-464 analogues. Bioorg. Med. Chem.21(4),874–882 (2012).
    • 68  Bai A, Szulc ZM, Bielawski J et al. Synthesis and bioevaluation of omega-N-amino analogs of B13. Bioorg. Med. Chem.17(5),1840–1848 (2009).
    • 69  Bhabak KP, Arenz C. Novel amide- and sulfonamide-based aromatic ethanolamines: effects of various substituents on the inhibition of acid and neutral ceramidases. Bioorg. Med. Chem.20(20),6162–6170 (2012).
    • 70  Abe A, Inokuchi J, Jimbo M et al. Improved inhibitors of glucosylceramide synthase. J. Biochem.111(2),191–196 (1992).
    • 71  Inokuchi J, Radin NS. Preparation of the active isomer of 1-phenyl-2-decanoylamino-3-morpholino-1-propanol, inhibitor of murine glucocerebroside synthetase. J. Lipid Res.28(5),565–571 (1987).
    • 72  Uemura K, Sugiyama E, Tamai C, Hara A, Taketomi T, Radin NS. Effect of an inhibitor of glucosylceramide synthesis on cultured rabbit skin fibroblasts. J. Biochem.108(4),525–530 (1990).
    • 73  Abe A, Radin NS, Shayman JA et al. Structural and stereochemical studies of potent inhibitors of glucosylceramide synthase and tumor cell growth. J. Lipid Res.36(3),611–621 (1995).
    • 74  Maurer BJ, Melton L, Billups C, Cabot MC, Reynolds CP. Synergistic cytotoxicity in solid tumor cell lines between N-(4-hydroxyphenyl)retinamide and modulators of ceramide metabolism. J. Natl Cancer Inst.92(23),1897–1909 (2000).
    • 75  Lee L, Abe A, Shayman JA. Improved inhibitors of glucosylceramide synthase. J. Biol. Chem.274(21),14662–14669 (1999).
    • 76  Hillaert U, Boldin-Adamsky S, Rozenski J, Busson R, Futerman AH, van Calenbergh S. Synthesis and biological evaluation of novel PDMP analogues. Bioorg. Med. Chem.14(15),5273–5284 (2006).
    • 77  Larsen SD, Wilson MW, Abe A et al. Property-based design of a glucosylceramide synthase inhibitor that reduces glucosylceramide in the brain. J. Lipid Res.53(2),282–291 (2012).
    • 78  Yatomi Y, Ruan F, Megidish T, Toyokuni T, Hakomori S, Igarashi Y. N,N-dimethylsphingosine inhibition of sphingosine kinase and sphingosine 1-phosphate activity in human platelets. Biochemistry35(2),626–633 (1996).
    • 79  Kohama T, Olivera A, Edsall L, Nagiec MM, Dickson R, Spiegel S. Molecular cloning and functional characterization of murine sphingosine kinase. J. Biol. Chem.273(37),23722–23728 (1998).
    • 80  Liu H, Sugiura M, Nava VE et al. Molecular cloning and functional characterization of a novel mammalian sphingosine kinase type 2 isoform. J. Biol. Chem.275(26),19513–19520 (2000).
    • 81  Shirahama T, Sweeney EA, Sakakura C et al.In vitro and in vivo induction of apoptosis by sphingosine and N, N-dimethylsphingosine in human epidermoid carcinoma KB-3–1 and its multidrug-resistant cells. Clin. Cancer Res.3(2),257–264 (1997).
    • 82  Sweeney EA, Sakakura C, Shirahama T et al. Sphingosine and its methylated derivative N,N-dimethylsphingosine (DMS) induce apoptosis in a variety of human cancer cell lines. Int. J. Cancer66(3),358–366 (1996).
    • 83  Igarashi Y, Hakomori S, Toyokuni T et al. Effect of chemically well-defined sphingosine and its N-methyl derivatives on protein kinase C and src kinase activities. Biochemistry28(17),6796–6800 (1989).
    • 84  Sugiura M, Kono K, Liu H et al. Ceramide kinase, a novel lipid kinase. Molecular cloning and functional characterization. J. Biol. Chem.277(26),23294–23300 (2002).
    • 85  Azuma H, Takahara S, Horie S, Muto S, Otsuki Y, Katsuoka Y. Induction of apoptosis in human bladder cancer cells in vitro and in vivo caused by FTY720 treatment. J. Urol.169(6),2372–2377 (2003).
    • 86  Ubai T, Azuma H, Kotake Y et al. FTY720 induced Bcl-associated and Fas-independent apoptosis in human renal cancer cells in vitro and significantly reduced in vivo tumor growth in mouse xenograft. Anticancer Res.27(1A),75–88 (2007).
    • 87  Shen Y, Cai M, Xia W et al. FTY720, a synthetic compound from Isaria sinclairii, inhibits proliferation and induces apoptosis in pancreatic cancer cells. Cancer Lett.254(2),288–297 (2007).
    • 88  Nagaoka Y, Otsuki K, Fujita T, Uesato S. Effects of phosphorylation of immunomodulatory agent FTY720 (fingolimod) on antiproliferative activity against breast and colon cancer cells. Biol. Pharm. Bull.31(6),1177–1181 (2008).
    • 89  Estrada-Bernal A, Palanichamy K, Ray Chaudhury A, Van Brocklyn JR. Induction of brain tumor stem cell apoptosis by FTY720: a potential therapeutic agent for glioblastoma. Neuro Oncol.14(4),405–415 (2012).
    • 90  Azuma H, Takahara S, Ichimaru N et al. Marked prevention of tumor growth and metastasis by a novel immunosuppressive agent, FTY720, in mouse breast cancer models. Cancer Res.62(5),1410–1419 (2002).
    • 91  Ho JW, Man K, Sun CK, Lee TK, Poon RT, Fan ST. Effects of a novel immunomodulating agent, FTY720, on tumor growth and angiogenesis in hepatocellular carcinoma. Mol. Cancer Ther.4(9),1430–1438 (2005).
    • 92  Matsuoka Y, Nagahara Y, Ikekita M, Shinomiya T. A novel immunosuppressive agent FTY720 induced AKT dephosphorylation in leukemia cells. Br. J. Pharmacol.138(7),1303–1312 (2003).
    • 93  Vessey DA, Kelley M, Zhang J, Li L, Tao R, Karliner JS. Dimethylsphingosine and FTY720 inhibit the SK1 form but activate the SK2 form of sphingosine kinase from rat heart. J. Biochem. Mol. Toxicol.21(5),273–279 (2007).
    • 94  Kharel Y, Lee S, Snyder AH et al. Sphingosine kinase 2 is required for modulation of lymphocyte traffic by FTY720. J. Biol. Chem.280(44),36865–36872 (2005).
    • 95  Zemann B, Kinzel B, Müller M et al. Sphingosine kinase type 2 is essential for lymphopenia induced by the immunomodulatory drug FTY720. Blood107(4),1454–1458 (2006).
    • 96  Brinkmann V, Davis MD, Heise CE et al. The immune modulator FTY720 targets sphingosine 1-phosphate receptors. J. Biol. Chem.277(24),21453–21457 (2002).
    • 97  Gräler MH, Goetzl EJ. The immunosuppressant FTY720 down-regulates sphingosine 1-phosphate G-protein-coupled receptors. FASEB J.18(3),551–553 (2004).
    • 98  Hamada M, Nakamura M, Kiuchi M et al. Removal of sphingosine 1-phosphate receptor-3 (S1P(3)) agonism is essential, but inadequate to obtain immunomodulating 2-aminopropane-1,3-diol S1P(1) agonists with reduced effect on heart rate. J. Med. Chem.53(8),3154–3168 (2010).
    • 99  Watson DG, Tonelli F, Al Osaimi M et al. The roles of sphingosine kinases 1 and 2 in regulating the Warburg effect in prostate cancer cells. Cell. Signal.25(4),1011–1017 (2013).
    • 100  Shimizu H, Takahashi M, Kaneko T et al. KRP-203, a novel synthetic immunosuppressant, prolongs graft survival and attenuates chronic rejection in rat skin and heart allografts. Circulation111(2),222–229 (2005).
    • 101  Fujishiro J, Kudou S, Iwai S et al. Use of sphingosine-1-phosphate 1 receptor agonist, KRP-203, in combination with a subtherapeutic dose of cyclosporine A for rat renal transplantation. Transplantation82(6),804–812 (2006).
    • 102  Bieberich E, Kawaguchi T, Yu RK. N-acylated serinol is a novel ceramide mimic inducing apoptosis in neuroblastoma cells. J. Biol. Chem.275(1),177–181 (2000).
    • 103  Loveridge C, Tonelli F, Leclercq T et al. The sphingosine kinase 1 inhibitor 2-(p-hydroxyanilino)-4-(p-chlorophenyl)thiazole induces proteasomal degradation of sphingosine kinase 1 in mammalian cells. J. Biol. Chem.285(50),38841–38852 (2010).
    • 104  Kono K, Tanaka M, Mizuno T, Kodama K, Ogita T, Kohama T. B-535a, b and c, new sphingosine kinase inhibitors, produced by a marine bacterium; taxonomy, fermentation, isolation, physico-chemical properties and structure determination. J. Antibiot. (Tokyo)53(8),753–758 (2000).
    • 105  Struckhoff AP, Bittman R, Burow ME et al. Novel ceramide analogs as potential chemotherapeutic agents in breast cancer. J. Pharmacol. Exp. Ther.309(2),523–532 (2004).▪ Introducing conjugated system into sphingosine backbone increasing apoptotic activity.
    • 106  De Jonghe S, Van Overmeire I, Gunst J et al. Synthesis and apoptogenic activity of fluorinated ceramide and dihydroceramide analogues. Bioorg. Med. Chem. Lett.9(21),3159–3164 (1999).
    • 107  Shikata K, Niiro H, Azuma H, Ogino K, Tachibana T. Apoptotic activities of C2-ceramide and C2-dihydroceramide homologues against HL-60 cells. Bioorg. Med. Chem.11(13),2723–2728 (2003).
    • 108  Shikata K, Niiro H, Azuma H, Tachibana T, Ogino K. Synthesis of non-natural C2-homo-ceramide and its apoptotic activity against HL-60 cells. Bioorg. Med. Chem. Lett.13(4),613–616 (2003).
    • 109  Niiro H, Azuma H, Tanago S et al. (3Z)-2-acetylamino-3-octadecen-1-ol as a potent apoptotic agent against HL-60 cells. Bioorg. Med. Chem.12(1),45–51 (2004).
    • 110  Bedia C, Canals D, Matabosch X et al. Cytotoxicity and acid ceramidase inhibitory activity of 2-substituted aminoethanol amides. Chem. Phys. Lipids156(1–2),33–40 (2008).
    • 111  Lu X, Arthur G, Bittman R. Synthesis of a novel ceramide analogue via Tebbe methylenation and evaluation of its antiproliferative activity. Org. Lett.7(8),1645–1648 (2005).
    • 112  Kang JH, Garg H, Sigano DM, Francella N, Blumenthal R, Marquez VE. Ceramides: branched alkyl chains in the sphingolipid siblings of diacylglycerol improve biological potency. Bioorg. Med. Chem.17(4),1498–1505 (2009).
    • 113  Singh A, Ha HJ, Park J, Kim JH, Lee WK. 3,4-disubstituted oxazolidin-2-ones as constrained ceramide analogs with anticancer activities. Bioorg. Med. Chem.19(21),6174–6181 (2011).
    • 114  Yasuda S, Kitagawa H, Ueno M et al. A novel inhibitor of ceramide trafficking from the endoplasmic reticulum to the site of sphingomyelin synthesis. J. Biol. Chem.276(47),43994–44002 (2001).
    • 115  Duriš A, Wiesenganger T, Moravcíková D et al. Expedient and practical synthesis of CERT-dependent ceramide trafficking inhibitor HPA-12 and its analogues. Org. Lett.13(7),1642–1645 (2011).
    • 116  Nakamura Y, Matsubara R, Kitagawa H et al. Stereoselective synthesis and structure-activity relationship of novel ceramide trafficking inhibitors. (1R,3R)-N-(3-hydroxy-1-hydroxymethyl-3-phenylpropyl)dodecanamide and its analogues. J. Med. Chem.46(17),3688–3695 (2003).
    • 117  Camacho L, Simbari F, Garrido M et al. 3-deoxy-3,4-dehydro analogs of XM462. Preparation and activity on sphingolipid metabolism and cell fate. Bioorg. Med. Chem.20(10),3173–3179 (2012).
    • 118  Novgorodov SA, Szulc ZM, Luberto C et al. Positively charged ceramide is a potent inducer of mitochondrial permeabilization. J. Biol. Chem.280(16),16096–16105 (2005).
    • 119  Kim S, Cho M, Lee T, Lee S, Min HY, Lee SK. Design, synthesis, and preliminary biological evaluation of a novel triazole analogue of ceramide. Bioorg. Med. Chem. Lett.17(16),4584–4587 (2007).
    • 120  Grijalvo S, Bedia C, Triola G et al. Design, synthesis and activity as acid ceramidase inhibitors of 2-oxooctanoyl and N-oleoylethanolamine analogues. Chem. Phys. Lipids144(1),69–84 (2006).
    • 121  Senkal CE, Ponnusamy S, Rossi MJ et al. Potent antitumor activity of a novel cationic pyridinium-ceramide alone or in combination with gemcitabine against human head and neck squamous cell carcinomas in vitro and in vivo.?. J. Pharmacol. Exp. Ther.317(3),1188–1199 (2006).
    • 122  Separovic D, Saad ZH, Edwin EA et al. C16-ceramide analog combined with Pc 4 photodynamic therapy evokes enhanced total ceramide accumulation, promotion of DEVDase activation in the absence of apoptosis, and augmented overall cell killing. J. Lipids doi:10.1155/2011/713867 (2011) (Epub ahead of print).
    • 123  Dindo D, Dahm F, Szulc Z et al. Cationic long-chain ceramide LCL-30 induces cell death by mitochondrial targeting in SW403 cells. Mol. Cancer Ther.5(6),1520–1529 (2006).
    • 124  Johnson CR, Chun J, Bittman R, Jarvis WD. Intrinsic cytotoxicity and chemomodulatory actions of novel phenethylisothiocyanate sphingoid base derivatives in HL-60 human promyelocytic leukemia cells. J. Pharmacol. Exp. Ther.309(2),452–461 (2004).
    • 125  Antoon JW, Liu J, Ponnapakkam AP, Gestaut MM, Foroozesh M, Beckman BS. Novel D: -erythroN-octanoyl sphingosine analogs as chemo- and endocrine-resistant breast cancer therapeutics. Cancer Chemother. Pharmacol.65(6),1191–1195 (2010).
    • 126  Kim JW, Kim YW, Inagaki Y et al. Synthesis and evaluation of sphingoid analogs as inhibitors of sphingosine kinases. Bioorg. Med. Chem.13(10),3475–3485 (2005).
    • 127  Oh JE, So KS, Lim SJ, Kim MY. Induction of apoptotic cell death by a ceramide analog in PC-3 prostate cancer cells. Arch. Pharm. Res.29(12),1140–1146 (2006).
    • 128  Wong L, Tan SS, Lam Y, Melendez AJ. Synthesis and evaluation of sphingosine analogues as inhibitors of sphingosine kinases. J. Med. Chem.52(12),3618–3626 (2009).
    • 129  Antoon JW, Liu J, Gestaut MM, Burow ME, Beckman BS, Foroozesh M. Design, synthesis, and biological activity of a family of novel ceramide analogues in chemoresistant breast cancer cells. J. Med. Chem.52(18),5748–5752 (2009).
    • 130  Liu J, Antoon JW, Ponnapakkam A, Beckman BS, Foroozesh M. Novel anti-viability ceramide analogs: design, synthesis, and structure-activity relationship studies of substituted (S)-2-(benzylideneamino)-3-hydroxy-N-tetradecylpropanamides. Bioorg. Med. Chem.18(14),5316–5322 (2010).
    • 131  Antoon JW, Beckman BS. Anti-proliferative effects of the novel ceramide analog (S)-2-(benzylideneamino)-3-hydroxy-N-tetrade-cylpropanamide in chemoresistant cancer. Bioorg. Med. Chem. Lett.22(7),2624–2628 (2012).
    • 132  Nichols CE, Lamb HK, Lockyer M et al. Characterization of Salmonella typhimurium YegS, a putative lipid kinase homologous to eukaryotic sphingosine and diacylglycerol kinases. Proteins68(1),13–25 (2007).
    • 133  Cho US, Xu W. Crystal structure of a protein phosphatase 2A heterotrimeric holoenzyme. Nature445(7123),53–57 (2007).
    • 134  Inoue T, Okino N, Kakuta Y et al. Mechanistic insights into the hydrolysis and synthesis of ceramide by neutral ceramidase. J. Biol. Chem.284(14),9566–9577 (2009).