Hostname: page-component-8448b6f56d-42gr6 Total loading time: 0 Render date: 2024-04-17T16:21:41.533Z Has data issue: false hasContentIssue false

Polyamines and cancer: implications for chemotherapy and chemoprevention

Published online by Cambridge University Press:  22 February 2013

Shannon L. Nowotarski
Affiliation:
Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA
Patrick M. Woster
Affiliation:
Department of Drug Discovery and Biomedical Sciences, The Medical University of South Carolina, Charleston, SC 29425, USA
Robert A. Casero Jr*
Affiliation:
Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA
*
*Corresponding author: Robert A. Casero, The Johns Hopkins University School of Medicine, Bunting/Blaustein Cancer Research Building 1, 1650 Orleans Street – Room 551, Baltimore, MD 21287, USA. E-mail: rcasero@jhmi.edu

Abstract

Polyamines are small organic cations that are essential for normal cell growth and development in eukaryotes. Under normal physiological conditions, intracellular polyamine concentrations are tightly regulated through a dynamic network of biosynthetic and catabolic enzymes, and a poorly characterised transport system. This precise regulation ensures that the intracellular concentration of polyamines is maintained within strictly controlled limits. It has frequently been observed that the metabolism of, and the requirement for, polyamines in tumours is frequently dysregulated. Elevated levels of polyamines have been associated with breast, colon, lung, prostate and skin cancers, and altered levels of rate-limiting enzymes in both biosynthesis and catabolism have been observed. Based on these observations and the absolute requirement for polyamines in tumour growth, the polyamine pathway is a rational target for chemoprevention and chemotherapeutics. Here we describe the recent advances made in the polyamine field and focus on the roles of polyamines and polyamine metabolism in neoplasia through a discussion of the current animal models for the polyamine pathway, chemotherapeutic strategies that target the polyamine pathway, chemotherapeutic clinical trials for polyamine pathway-specific drugs and ongoing clinical trials targeting polyamine biosynthesis.

Type
Review Article
Copyright
Copyright © Cambridge University Press 2013 

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

1Pegg, A.E. (1988) Polyamine metabolism and its importance in neoplastic growth and a target for chemotherapy. Cancer Research 48, 759-774Google Scholar
2Gerner, E.W. and Meyskens, F.L. Jr (2004) Polyamines and cancer: old molecules, new understanding. Nature Reviews. Cancer 4, 781-792CrossRefGoogle ScholarPubMed
3Wallace, H.M., Fraser, A.V. and Hughes, A. (2003) A perspective of polyamine metabolism. Biochemical Journal 376(Pt 1), 1-14Google Scholar
4Matthews, H.R. (1993) Polyamines, chromatin structure and transcription. Bioessays 15, 561-566CrossRefGoogle ScholarPubMed
5Feuerstein, B.G. et al. (1991) Implications and concepts of polyamine-nucleic acid interactions. Journal of Cellular Biochemistry 46, 37-47Google Scholar
6Ha, H.C. et al. (1998) The natural polyamine spermine functions directly as a free radical scavenger. Proceedings of the National Academy of Sciences of the United States of America 95, 11140-11145Google Scholar
7Kurata, H.T., Marton, L.J. and Nichols, C.G. (2006) The polyamine binding site in inward rectifier K+ channels. Journal of General Physiology 127, 467-480Google Scholar
8Schuber, F. (1989) Influence of polyamines on membrane functions. Biochemical Journal 260, 1-10CrossRefGoogle ScholarPubMed
9Casero, R.A. Jr and Marton, L.J. (2007) Targeting polyamine metabolism and function in cancer and other hyperproliferative diseases. Nature Reviews. Drug Discovery 6, 373-390Google Scholar
10Pegg, A.E. (2009) Mammalian polyamine metabolism and function. IUBMB Life 61, 880-894Google Scholar
11Wallace, H.M. (2000) The physiological role of the polyamines. European Journal of Clinical Investigation 30, 1-3Google Scholar
12Russell, D. and Snyder, S.H. (1968) Amine synthesis in rapidly growing tissues: ornithine decarboxylase activity in regenerating rat liver, chick embryo, and various tumors. Proceedings of the National Academy of Sciences of the United States of America 60, 1420-1427Google Scholar
13Manni, A. et al. (1995) Involvement of the polyamine pathway in breast cancer progression. Cancer Letters 92, 49-57CrossRefGoogle ScholarPubMed
14Gupta, S. et al. (2000) Chemoprevention of prostate carcinogenesis by alpha-difluoromethylornithine in TRAMP mice. Cancer Research 60, 5125-5133Google ScholarPubMed
15Gilmour, S.K. (2007) Polyamines and nonmelanoma skin cancer. Toxicology and Applied Pharmacology 224, 249-256Google Scholar
16Upp, J.R. Jr, et al. (1988) Polyamine levels and gastrin receptors in colon cancers. Annals of Surgery 207, 662-669Google Scholar
17Tobias, K.E., Shor, J. and Kahana, C. (1995) c-Myc and Max transregulate the mouse ornithine decarboxylase promoter through interaction with two downstream CACGTG motifs. Oncogene 11, 1721-1727Google Scholar
18Shantz, L.M. and Levin, V.A. (2007) Regulation of ornithine decarboxylase during oncogenic transformation: mechanisms and therapeutic potential. Amino Acids 33, 213-223CrossRefGoogle ScholarPubMed
19Holtta, E., Sistonen, L. and Alitalo, K. (1988) The mechanisms of ornithine decarboxylase deregulation in c-Ha-ras oncogene-transformed NIH 3T3 cells. Journal of Biological Chemistry 263, 4500-4507Google Scholar
20Ignatenko, N.A. et al. (2004) Suppression of polyamine catabolism by activated Ki-ras in human colon cancer cells. Molecular Carcinogenesis 39, 91-102Google Scholar
21Shantz, L.M. (2004) Transcriptional and translational control of ornithine decarboxylase during Ras transformation. Biochemical Journal 377(Pt 1), 257-264Google Scholar
22Pegg, A.E. (2006) Regulation of ornithine decarboxylase. Journal of Biological Chemistry 281, 14529-14532Google Scholar
23Nowotarski, S.L. and Shantz, L.M. (2010) Cytoplasmic accumulation of the RNA-binding protein HuR stabilizes the ornithine decarboxylase transcript in a murine nonmelanoma skin cancer model. Journal of Biological Chemistry 285, 31885-31894Google Scholar
24Pendeville, H. et al. (2001) The ornithine decarboxylase gene is essential for cell survival during early murine development. Molecular and Cellular Biology 21, 6549-6558Google Scholar
25Katz, A. and Kahana, C. (1987) Transcriptional activation of mammalian ornithine decarboxylase during stimulated growth. Molecular and Cellular Biology 7, 2641-2643Google Scholar
26Verma, A.K., Hsieh, J.T. and Pong, R.C. (1988) Mechanisms involved in ornithine decarboxylase induction by 12-O-tetradecanoylphorbol-13-acetate, a potent mouse skin tumor promoter and an activator of protein kinase C. Advances in Experimental Medicine and Biology 250, 273-290Google Scholar
27Abrahamsen, M.S. et al. (1992) Multiple DNA elements responsible for transcriptional regulation of the ornithine decarboxylase gene by protein kinase A. Journal of Biological Chemistry 267, 18866-18873Google Scholar
28Zhao, B. and Butler, A.P. (2001) Core promoter involvement in the induction of rat ornithine decarboxylase by phorbol esters. Molecular Carcinogenesis 32, 92-99CrossRefGoogle ScholarPubMed
29Bello-Fernandez, C., Packham, G. and Cleveland, J.L. (1993) The ornithine decarboxylase gene is a transcriptional target of c-Myc. Proceedings of the National Academy of Sciences of the United States of America 90, 7804-7808CrossRefGoogle ScholarPubMed
30Packham, G. and Cleveland, J.L. (1995) The role of ornithine decarboxylase in c-Myc-induced apoptosis. Current Topics in Microbiology and Immunology 194, 283-290Google Scholar
31Murakami, Y. et al. (1992) Ornithine decarboxylase is degraded by the 26S proteasome without ubiquitination. Nature 360, 597-599Google Scholar
32Nilsson, J. et al. (1997) Polyamines regulate both transcription and translation of the gene encoding ornithine decarboxylase antizyme in mouse. European Journal of Biochemistry/FEBS 250, 223-231CrossRefGoogle Scholar
33Matsufuji, S. et al. (1995) Autoregulatory frameshifting in decoding mammalian ornithine decarboxylase antizyme. Cell 80, 51-60CrossRefGoogle ScholarPubMed
34Stanley, B.A., Pegg, A.E. and Holm, I. (1989) Site of pyruvate formation and processing of mammalian S-adenosylmethionine decarboxylase proenzyme. Journal of Biological Chemistry 264, 21073-21079Google Scholar
35Ikeguchi, Y., Bewley, M.C. and Pegg, A.E. (2006) Aminopropyltransferases: function, structure and genetics. Journal of Biochemistry 139, 1-9Google Scholar
36Korhonen, V.P. et al. (1995) Molecular cloning of a cDNA encoding human spermine synthase. DNA and Cell Biology 14, 841-847Google Scholar
37Wahlfors, J. et al. (1990) Human spermidine synthase: cloning and primary structure. DNA and Cell Biology 9, 103-110Google Scholar
38Casero, R.A. Jr and Pegg, A.E. (1993) Spermidine/spermine N1-acetyltransferase – the turning point in polyamine metabolism. FASEB Journal 7, 653-661CrossRefGoogle ScholarPubMed
39Casero, R.A. and Pegg, A.E. (2009) Polyamine catabolism and disease. Biochemical Journal 421, 323-338Google Scholar
40Xie, X., Gillies, R.J. and Gerner, E.W. (1997) Characterization of a diamine exporter in Chinese hamster ovary cells and identification of specific polyamine substrates. Journal of Biological Chemistry 272, 20484-20489CrossRefGoogle ScholarPubMed
41Vujcic, S. et al. (2003) Genomic identification and biochemical characterization of the mammalian polyamine oxidase involved in polyamine back-conversion. Biochemical Journal 370(Pt 1), 19-28CrossRefGoogle ScholarPubMed
42Wu, T., Yankovskaya, V. and McIntire, W.S. (2003) Cloning, sequencing, and heterologous expression of the murine peroxisomal flavoprotein, N1-acetylated polyamine oxidase. Journal of Biological Chemistry 278, 20514-20525Google Scholar
43Wang, Y. et al. (2001) Cloning and characterization of a human polyamine oxidase that is inducible by polyamine analogue exposure. Cancer Research 61, 5370-5373Google Scholar
44Vujcic, S. et al. (2002) Identification and characterization of a novel flavin-containing spermine oxidase of mammalian cell origin. Biochemical Journal 367(Pt 3), 665-675Google Scholar
45Wang, Y. et al. (2003) Properties of purified recombinant human polyamine oxidase, PAOh1/SMO. Biochemical and Biophysical Research Communications 304, 605-611Google Scholar
46Babbar, N. and Casero, R.A. Jr (2006) Tumor necrosis factor-alpha increases reactive oxygen species by inducing spermine oxidase in human lung epithelial cells: a potential mechanism for inflammation-induced carcinogenesis. Cancer Research 66, 11125-11130Google Scholar
47Goodwin, A.C. et al. (2011) Polyamine catabolism contributes to enterotoxigenic Bacteroides fragilis-induced colon tumorigenesis. Proceedings of the National Academy of Sciences of the United States of America 108, 15354-15359Google Scholar
48Bardocz, S. et al. (1995) The importance of dietary polyamines in cell regeneration and growth. British Journal of Nutrition 73, 819-828Google Scholar
49Kahana, C. (2009) Regulation of cellular polyamine levels and cellular proliferation by antizyme and antizyme inhibitor. Essays in Biochemistry 46, 47-61Google Scholar
50Poulin, R., Casero, R.A. and Soulet, D. (2012) Recent advances in the molecular biology of metazoan polyamine transport. Amino Acids 42, 711-723Google Scholar
51Lessard, M. et al. (1995) Hormonal and feedback regulation of putrescine and spermidine transport in human breast cancer cells. Journal of Biological Chemistry 270, 1685-1694Google Scholar
52Mitchell, J.L. et al. (2004) Antizyme induction mediates feedback limitation of the incorporation of specific polyamine analogues in tissue culture. Biochemical Journal 384(Pt 2), 271-279Google Scholar
53Soulet, D. et al. (2004) A fluorescent probe of polyamine transport accumulates into intracellular acidic vesicles via a two-step mechanism. Journal of Biological Chemistry 279, 49355-49366Google Scholar
54Belting, M. et al. (2003) Glypican-1 is a vehicle for polyamine uptake in mammalian cells: a pivotal role for nitrosothiol-derived nitric oxide. Journal of Biological Chemistry 278, 47181-47189Google Scholar
55Roy, U.K. et al. (2008) Activated K-RAS increases polyamine uptake in human colon cancer cells through modulation of caveolar endocytosis. Molecular Carcinogenesis 47, 538-553Google Scholar
56Uemura, T. et al. (2010) Polyamine transport is mediated by both endocytic and solute carrier transport mechanisms in the gastrointestinal tract. American Journal of Physiology. Gastrointestinal and Liver Physiology 299, G517-G522Google Scholar
57Casero, R.A. Jr, et al. (2003) The role of polyamine catabolism in anti-tumour drug response. Biochemical Society Transactions 31, 361-365Google Scholar
58Giardiello, F.M. et al. (1997) Ornithine decarboxylase and polyamines in familial adenomatous polyposis. Cancer Research 57, 199-201Google Scholar
59Martinez, M.E. et al. (2003) Pronounced reduction in adenoma recurrence associated with aspirin use and a polymorphism in the ornithine decarboxylase gene. Proceedings of the National Academy of Sciences of the United States of America 100, 7859-7864Google Scholar
60Elmets, C.A. and Athar, M. (2010) Targeting ornithine decarboxylase for the prevention of nonmelanoma skin cancer in humans. Cancer Prevention Research (Philadelphia, PA) 3, 8-11Google Scholar
61Manni, A. et al. (1996) Prognostic influence on survival of increased ornithine decarboxylase activity in human breast cancer. Clinical Cancer Research 2, 1901-1906Google ScholarPubMed
62Chaturvedi, R. et al. (2011) Spermine oxidase mediates the gastric cancer risk associated with Helicobacter pylori CagA. Gastroenterology 141, 1696-1708 e1-2Google Scholar
63Xu, H. et al. (2004) Spermine oxidation induced by Helicobacter pylori results in apoptosis and DNA damage: implications for gastric carcinogenesis. Cancer Research 64, 8521-8525Google Scholar
64Goodwin, A.C. et al. (2008) Increased spermine oxidase expression in human prostate cancer and prostatic intraepithelial neoplasia tissues. Prostate 68, 766-772Google Scholar
65Hong, S.K. et al. (2010) Increased expression and cellular localization of spermine oxidase in ulcerative colitis and relationship to disease activity. Inflammatory Bowel Diseases 16, 1557-1566Google Scholar
66Alhonen, L. et al. (1995) Life-long over-expression of ornithine decarboxylase (ODC) gene in transgenic mice does not lead to generally enhanced tumorigenesis or neuronal degeneration. International Journal of Cancer 63, 402-404Google Scholar
67Guo, Y. et al. (1999) Conversion of C57Bl/6 mice from a tumor promotion-resistant to a – sensitive phenotype by enhanced ornithine decarboxylase expression. Molecular Carcinogenesis 26, 32-36Google Scholar
68Guo, Y., Cleveland, J.L. and O'Brien, T.G. (2005) Haploinsufficiency for odc modifies mouse skin tumor susceptibility. Cancer Research 65, 1146-1149CrossRefGoogle ScholarPubMed
69Nilsson, J.A. et al. (2005) Targeting ornithine decarboxylase in Myc-induced lymphomagenesis prevents tumor formation. Cancer Cell 7, 433-444Google Scholar
70Heljasvaara, R. et al. (1997) Transgenic mice overexpressing ornithine and S-adenosylmethionine decarboxylases maintain a physiological polyamine homoeostasis in their tissues. Biochemical Journal 323(Pt 2), 457-462CrossRefGoogle ScholarPubMed
71Shi, C. et al. (2012) S-adenosylmethionine decarboxylase overexpression inhibits mouse skin tumor promotion. Carcinogenesis 33, 1310-1318Google Scholar
72Scuoppo, C. et al. (2012) A tumour suppressor network relying on the polyamine-hypusine axis. Nature 487, 244-248Google Scholar
73Shi, C. et al. (2012) Characterization of transgenic mice with overexpression of spermidine synthase. Amino Acids 42, 495-505Google Scholar
74Kauppinen, L. et al. (1993) Transgenic mice over-expressing the human spermidine synthase gene. Biochemical Journal 293(Pt 2), 513-516Google Scholar
75Welsh, P.A. et al. (2012) Spermine synthase overexpression in vivo does not increase susceptibility to DMBA/TPA skin carcinogenesis or Min-Apc intestinal tumorigenesis. Cancer Biology and Therapy 13, 358-368Google Scholar
76Feith, D.J., Shantz, L.M. and Pegg, A.E. (2001) Targeted antizyme expression in the skin of transgenic mice reduces tumor promoter induction of ornithine decarboxylase and decreases sensitivity to chemical carcinogenesis. Cancer Research 61, 6073-6081Google Scholar
77Fong, L.Y., Feith, D.J. and Pegg, A.E. (2003) Antizyme overexpression in transgenic mice reduces cell proliferation, increases apoptosis, and reduces N-nitrosomethylbenzylamine-induced forestomach carcinogenesis. Cancer Research 63, 3945-3954Google Scholar
78Pietila, M. et al. (2001) Relation of skin polyamines to the hairless phenotype in transgenic mice overexpressing spermidine/spermine N-acetyltransferase. Journal of Investigative Dermatology 116, 801-805Google Scholar
79Coleman, C.S. et al. (2002) Targeted expression of spermidine/spermine N1-acetyltransferase increases susceptibility to chemically induced skin carcinogenesis. Carcinogenesis 23, 359-364Google Scholar
80Kee, K. et al. (2004) Activated polyamine catabolism depletes acetyl-CoA pools and suppresses prostate tumor growth in TRAMP mice. Journal of Biological Chemistry 279, 40076-40083Google Scholar
81Tucker, J.M. et al. (2005) Potent modulation of intestinal tumorigenesis in Apcmin/+ mice by the polyamine catabolic enzyme spermidine/spermine N1-acetyltransferase. Cancer Research 65, 5390-5398Google Scholar
82Weeks, C.E. et al. (1982) Alpha-difluoromethylornithine, an irreversible inhibitor of ornithine decarboxylase, inhibits tumor promoter-induced polyamine accumulation and carcinogenesis in mouse skin. Proceedings of the National Academy of Sciences of the United States of America 79, 6028-6032Google Scholar
83Prakash, N.J. et al. (1978) Effect of alpha-difluoromethylornithine, an enzyme-activated irreversible inhibitor of ornithine decarboxylase, on L1210 leukemia in mice. Cancer Research 38, 3059-3062Google Scholar
84Mamont, P.S. et al. (1978) Anti-proliferative properties of DL-alpha-difluoromethyl ornithine in cultured cells. A consequence of the irreversible inhibition of ornithine decarboxylase. Biochemical and Biophysical Research Communications 81, 58-66Google Scholar
85Luk, G.D. et al. (1981) Polyamines are necessary for the survival of human small-cell lung carcinoma in culture. Proceedings of the National Academy of Sciences of the United States of America 78, 2355-2358Google Scholar
86Luk, G.D. et al. (1983) Successful treatment with DL-alpha-difluoromethylornithine in established human small cell variant lung carcinoma implants in athymic mice. Cancer Research 43, 4239-4243Google ScholarPubMed
87Abeloff, M.D. et al. (1986) Phase II trials of alpha-difluoromethylornithine, an inhibitor of polyamine synthesis, in advanced small cell lung cancer and colon cancer. Cancer Treatment Reports 70, 843-845Google Scholar
88Abeloff, M.D. et al. (1984) Phase I trial and pharmacokinetic studies of alpha-difluoromethylornithine – an inhibitor of polyamine biosynthesis. Journal of Clinical Oncology 2, 124-130Google Scholar
89Mihich, E. (1963) Current studies with methylglyoxal-Bis(Guanylhydrazone). Cancer Research 23, 1375-1389Google Scholar
90Williams-Ashman, H.G. and Schenone, A. (1972) Methyl glyoxal bis(guanylhydrazone) as a potent inhibitor of mammalian and yeast S-adenosylmethionine decarboxylases. Biochemical and Biophysical Research Communications 46, 288-295Google Scholar
91Nass, M.M. (1984) Analysis of methylglyoxal bis(guanylhydrazone)-induced alterations of hamster tumor mitochondria by correlated studies of selective rhodamine binding, ultrastructural damage, DNA replication, and reversibility. Cancer Research 44, 2677-2688Google Scholar
92Secrist, J.A. 3rd (1987) New substrate analogues as inhibitors of S-adenosylmethionine decarboxylase. Nucleosides and Nucleotides 6, 73-83Google Scholar
93Wu, Y. and Woster, P.M. (1992) S-(5′-deoxy-5′-adenosyl)-1-ammonio-4-(methylsulfonio)-2-cyclopentene: a potent, enzyme-activated irreversible inhibitor of S-adenosylmethionine decarboxylase. Journal of Medicinal Chemistry 35, 3196-3201Google Scholar
94Danzin, C., Marchal, P. and Casara, P. (1990) Irreversible inhibition of rat S-adenosylmethionine decarboxylase by 5′-([(Z)-4-amino-2-butenyl]methylamino)-5′-deoxyadenosine. Biochemical Pharmacology 40, 1499-1503Google Scholar
95Barker, R.H. Jr, et al. (2009) Novel S-adenosylmethionine decarboxylase inhibitors for the treatment of human African trypanosomiasis. Antimicrobial Agents and Chemotherapy 53, 2052-2058Google Scholar
96Byers, T.L. et al. (1993) Effects of chronic 5′-([(Z)-4-amino-2-butenyl]methylamino)-5′-deoxy- adenosine (AbeAdo) treatment on polyamine and eIF-5A metabolism in AbeAdo-sensitive and -resistant L1210 murine leukaemia cells. Biochemical Journal 290(Pt 1), 115-121CrossRefGoogle Scholar
97Eskens, F.A. et al. (2000) Phase I and pharmacological study of weekly administration of the polyamine synthesis inhibitor SAM 486A (CGP 48 664) in patients with solid tumors. European organization for research and treatment of cancer early clinical studies group. Clinical Cancer Research 6, 1736-1743Google Scholar
98Zhou, H. et al. (2000) Population pharmacokinetics/toxicodynamics (PK/TD) relationship of SAM486A in phase I studies in patients with advanced cancers. Journal of Clinical Pharmacology 40, 275-283Google Scholar
99Paridaens, R. et al. (2000) A phase I study of a new polyamine biosynthesis inhibitor, SAM486A, in cancer patients with solid tumours. British Journal of Cancer 83, 594-601Google Scholar
100Pless, M. et al. (2004) Clinical efficacy, tolerability, and safety of SAM486A, a novel polyamine biosynthesis inhibitor, in patients with relapsed or refractory non-Hodgkin's lymphoma: results from a phase II multicenter study. Clinical Cancer Research 10, 1299-1305Google Scholar
101Regenass, U. et al. (1994) CGP 48664, a new S-adenosylmethionine decarboxylase inhibitor with broad spectrum antiproliferative and antitumor activity. Cancer Research 54, 3210-3217Google Scholar
102Pegg, A.E., Tang, K.C. and Coward, J.K. (1982) Effects of S-adenosyl-1,8-diamino-3-thiooctane on polyamine metabolism. Biochemistry 21, 5082-5089Google Scholar
103Pegg, A.E. et al. (1989) Effect of S-adenosyl-1,12-diamino-3-thio-9-azadodecane, a multisubstrate adduct inhibitor of spermine synthase, on polyamine metabolism in mammalian cells. Biochemistry 28, 8446-8453Google Scholar
104Porter, C.W. and Bergeron, R.J. (1988) Regulation of polyamine biosynthetic activity by spermidine and spermine analogs – a novel antiproliferative strategy. Advances in Experimental Medicine and Biology 250, 677-690Google Scholar
105Casero, R.A. Jr and Woster, P.M. (2001) Terminally alkylated polyamine analogues as chemotherapeutic agents. Journal of Medicinal Chemistry 44, 1-26Google Scholar
106Casero, R.A. Jr and Woster, P.M. (2009) Recent advances in the development of polyamine analogues as antitumor agents. Journal of Medicinal Chemistry 52, 4551-4573Google Scholar
107Ha, H.C. et al. (1997) The role of polyamine catabolism in polyamine analogue-induced programmed cell death. Proceedings of the National Academy of Sciences of the United States of America 94, 11557-11562Google Scholar
108Pledgie, A. et al. (2005) Spermine oxidase SMO(PAOh1), Not N1-acetylpolyamine oxidase PAO, is the primary source of cytotoxic H2O2 in polyamine analogue-treated human breast cancer cell lines. Journal of Biological Chemistry 280, 39843-39851Google Scholar
109Casero, R.A. Jr, Gabrielson, E.W. and Pegg, A.E. (1994) Immunohistochemical staining of human spermidine/spermine N1-acetyltransferase superinduced in response to treatment with antitumor polyamine analogues. Cancer Research 54, 3955-3958Google Scholar
110Gabrielson, E.W., Pegg, A.E. and Casero, R.A. Jr (1999) The induction of spermidine/spermine N1-acetyltransferase (SSAT) is a common event in the response of human primary non-small cell lung carcinomas to exposure to the new antitumor polyamine analogue N1,N11-bis(ethyl)norspermine. Clinical Cancer Research 5, 1638-1641Google Scholar
111Bernacki, R.J. et al. (1995) Preclinical antitumor efficacy of the polyamine analogue N1, N11-diethylnorspermine administered by multiple injection or continuous infusion. Clinical Cancer Research 1, 847-857Google Scholar
112Hahm, H.A. et al. (2002) Phase I study of N(1),N(11)-diethylnorspermine in patients with non-small cell lung cancer. Clinical Cancer Research 8, 684-690Google Scholar
113Streiff, R.R. and Bender, J.F. (2001) Phase 1 study of N1-N11-diethylnorspermine (DENSPM) administered TID for 6 days in patients with advanced malignancies. Investigational New Drugs 19, 29-39Google Scholar
114Creaven, P.J. et al. (1997) Unusual central nervous system toxicity in a phase I study of N1N11 diethylnorspermine in patients with advanced malignancy. Investigational New Drugs 15, 227-234Google Scholar
115Davidson, N.E. et al. (1999) Clinical aspects of cell death in breast cancer: the polyamine pathway as a new target for treatment. Endocrine-Related Cancer 6, 69-73Google Scholar
116Wolff, A.C. et al. (2003) A phase II study of the polyamine analog N1,N11-diethylnorspermine (DENSpm) daily for five days every 21 days in patients with previously treated metastatic breast cancer. Clinical Cancer Research 9(Pt 1), 5922-5928Google Scholar
117Hector, S. et al. (2004) Polyamine catabolism in platinum drug action: interactions between oxaliplatin and the polyamine analogue N1,N11-diethylnorspermine at the level of spermidine/spermine N1-acetyltransferase. Molecular Cancer Therapeutics 3, 813-822Google Scholar
118Choi, W. et al. (2005) Combination of 5-fluorouracil and N1,N11-diethylnorspermine markedly activates spermidine/spermine N1-acetyltransferase expression, depletes polyamines, and synergistically induces apoptosis in colon carcinoma cells. Journal of Biological Chemistry 280, 3295-3304Google Scholar
119Pledgie-Tracy, A. et al. (2010) The role of the polyamine catabolic enzymes SSAT and SMO in the synergistic effects of standard chemotherapeutic agents with a polyamine analogue in human breast cancer cell lines. Cancer Chemotherapy and Pharmacology 65, 1067-1081Google Scholar
120Casero, R.A. Jr, et al. (1995) Growth and biochemical effects of unsymmetrically substituted polyamine analogues in human lung tumor cells 1. Cancer Chemotherapy and Pharmacology 36, 69-74Google Scholar
121Varghese, S. et al. (2008) Polyaminohydroxamic acids and polyaminobenzamides as isoform selective histone deacetylase inhibitors. Journal of Medicinal Chemistry 51, 2447-2456Google Scholar
122Varghese, S. et al. (2005) Alkyl-substituted polyaminohydroxamic acids: a novel class of targeted histone deacetylase inhibitors. Journal of Medicinal Chemistry 48, 6350-6365Google Scholar
123Wu, Y. et al. (2012) Oligoamine analogues in combination with 2-difluoromethylornithine synergistically induce re-expression of aberrantly silenced tumour-suppressor genes. Biochemical Journal 442, 693-701Google Scholar
124Huang, Y. et al. (2007) Inhibition of lysine-specific demethylase 1 by polyamine analogues results in reexpression of aberrantly silenced genes. Proceedings of the National Academy of Sciences of the United States of America 104, 8023-8028Google Scholar
125Carew, J.S. et al. (2008) The novel polyamine analogue CGC-11093 enhances the antimyeloma activity of bortezomib. Cancer Research 68, 4783-4790Google Scholar
126Hacker, A. et al. (2008) In vitro and in vivo effects of the conformationally restricted polyamine analogue CGC-11047 on small cell and non-small cell lung cancer cells. Cancer Chemotherapy and Pharmacology 63, 45-53Google Scholar
127Kuo, W.L. et al. (2009) A systems analysis of the chemosensitivity of breast cancer cells to the polyamine analogue PG-11047. BMC Medicine [Electronic Resource] 7, 77Google Scholar
128Ignatenko, N.A. et al. (2009) Gene expression analysis of HCT116 colon tumor-derived cells treated with the polyamine analog PG-11047. Cancer Genomics and Proteomics 6, 161-175Google Scholar
129Dredge, K. et al. (2009) The polyamine analog PG11047 potentiates the antitumor activity of cisplatin and bevacizumab in preclinical models of lung and prostate cancer. Cancer Chemotherapy and Pharmacology 65, 191-195Google Scholar
130Osland, A. and Kleppe, K. (1977) Polyamine induced aggregation of DNA. Nucleic Acids Research 4, 685-695Google Scholar
131Valasinas, A. et al. (2003) Long-chain polyamines (oligoamines) exhibit strong cytotoxicities against human prostate cancer cells. Bioorganic and Medicinal Chemistry 11, 4121-4131Google Scholar
132Huang, Y. et al. (2003) A novel polyamine analog inhibits growth and induces apoptosis in human breast cancer cells. Clinical Cancer Research 9, 2769-2777Google Scholar
133Marks, P. et al. (2001) Histone deacetylases and cancer: causes and therapies. Nature Reviews. Cancer 1, 194-202Google Scholar
134Arrowsmith, C.H. et al. (2012) Epigenetic protein families: a new frontier for drug discovery. Nature Reviews. Drug Discovery 11, 384-400Google Scholar
135Shi, Y. et al. (2004) Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119, 941-953Google Scholar
136Federico, R. et al. (2001) Inhibition of pig liver and Zea mays L. polyamine oxidase: a comparative study. Journal of Enzyme Inhibition 16, 147-155Google Scholar
137Bianchi, M. et al. (2006) Inhibition of polyamine and spermine oxidases by polyamine analogues. FEBS Journal 273, 1115-1123Google Scholar
138Sharma, S.K. et al. (2010) (Bis)urea and (bis)thiourea inhibitors of lysine-specific demethylase 1 as epigenetic modulators. Journal of Medicinal Chemistry 53, 5197-5212Google Scholar
139Sharma, S.K. et al. (2012) Polyamine-based small molecule epigenetic modulators. Medicinal Chemistry Communications 3, 14-21Google Scholar
140Huang, Y. et al. (2009) Novel oligoamine analogues inhibit lysine-specific demethylase 1 and induce reexpression of epigenetically silenced genes. Clinical Cancer Research 15, 7217-7228Google Scholar
141Hazeldine, S. et al. (2012) Low molecular weight amidoximes that act as potent inhibitors of lysine-specific demethylase 1. Journal of Medicinal Chemistry 55, 7378-7391Google Scholar
142Tsuji, T. et al. (2001) Induction of epithelial differentiation and DNA demethylation in hamster malignant oral keratinocyte by ornithine decarboxylase antizyme. Oncogene 20, 24-33Google Scholar
143Yamamoto, D. et al. (2010) Ornithine decarboxylase antizyme induces hypomethylation of genome DNA and histone H3 lysine 9 dimethylation (H3K9me2) in human oral cancer cell line. PLoS ONE 5, e12554Google Scholar
144Meyskens, F.L. Jr, et al. (1998) Effect of alpha-difluoromethylornithine on rectal mucosal levels of polyamines in a randomized, double-blinded trial for colon cancer prevention. Journal of the National Cancer Institute 90, 1212-1218Google Scholar
145Meyskens, F.L. Jr, et al. (2008) Difluoromethylornithine plus sulindac for the prevention of sporadic colorectal adenomas: a randomized placebo-controlled, double-blind trial. Cancer Prevention Research (Philadelphia, PA) 1, 32-38Google Scholar
146Huang, L. et al. (2010) Phospho-sulindac (OXT-922) inhibits the growth of human colon cancer cell lines: a redox/polyamine-dependent effect. Carcinogenesis 31, 1982-1990Google Scholar
147Mackenzie, G.G. et al. (2010) Phospho-sulindac (OXT-328), a novel sulindac derivative, is safe and effective in colon cancer prevention in mice. Gastroenterology 139, 1320-1332Google Scholar
148Bailey, H.H. (2010) A randomized, double-blind, placebo controlled phase 3 skin cancer prevention study of α-difluoromethylornithine in subjects with previous history of skin cancer. Cancer Prevention Research (Philadelphia, PA) 3, 35-47Google Scholar
149Alberts, D.S. et al. (2000) Chemoprevention of human actinic keratoses by topical 2-(difluoromethyl)-dl-ornithine. Cancer Epidemiology, Biomarkers and Prevention 9, 1281-1286Google Scholar
150Sinicrope, F.A. et al. (2011) Evaluation of difluoromethylornithine for the chemoprevention of Barrett's esophagus and mucosal dysplasia. Cancer Prevention Research 4, 829-839Google Scholar
151Simoneau, A.R. et al. (2008) The effect of difluoromethylornithine on decreasing prostate size and polyamines in men: results of a year-long phase IIb randomized placebo-controlled chemoprevention trial. Cancer Epidemiology, Biomarkers and Prevention 17, 292-299Google Scholar
152Babbar, N., Murray-Stewart, T. and Casero, R.A. Jr (2007) Inflammation and polyamine catabolism: the good, the bad and the ugly. Biochemical Society Transactions 35(Pt 2), 300-304Google Scholar
153Holley, J.L. et al. (1992) Targeting of tumor cells and DNA by a chlorambucil-spermidine conjugate. Cancer Research 52, 4190-4195Google Scholar
154Phanstiel, O.I. et al. (2000) The effect of polyamine homologation on the transport and cytotoxicity properties of polyamine-(DNA-intercalator) conjugates. Journal of Organic Chemistry 65, 7710Google Scholar
155Wang, C. et al. (2003) Molecular requirements for targeting the polyamine transport system. Synthesis and biological evaluation of polyamine-anthracene conjugates. Journal of Medicinal Chemistry 46, 2672-282Google Scholar

Further reading, resources and contacts

Here are journal review articles that were not featured above which provide useful information on the topic of polyamines.

Pegg, A.E. (2008) Spermidine/spermine-N(1)-acetyltransferase: a key metabolic regulator. American Journal of Physiology. Endocrinology and Metabolism 294, E995-E1010.

Wallace, H.M. and Niiranen, K. (2007) Polyamine analogues – an update. Amino Acids 33, 261-265.

Wallace, H.M. (2009) The polyamines: past, present and future. Essays in Biochemistry 46, 1-9.

Wang, Y. and Casero, R.A. (2006) Mammalian polyamine catabolism: a therapeutic target, a pathological problem, or both?. Journal of Biochemistry (Tokyo) 139, 17-25.

Seiler, N. (2003) Thirty years of polyamine-related approaches to cancer therapy. Retrospect and prospect. Part 2. Structural analogues and derivatives. Current Drug Targets 4, 565-585.

Here are textbooks that were not cited in the above article. These provide useful information concerning polyamines, methods for studying the polyamine pathway and the use of polyamines in drug discovery.

Cohen, S.S. (1998) A Guide to the Polyamines, Oxford University Press, New York, USA.

Pegg, A.E. and Casero, R.A. (2011) Polyamines: Methods and Protocols, Humana Press/Springer Science + Business Media, New York, USA.

Wallace, H.M. (2009) The Polyamines: Small Molecules in the Omics Era, Portland Press, London, UK.

Wang, J.Y. and Casero, R.A. (2006) Polyamine Cell Signaling: Physiology, Pharmacology, and Cancer Research, Humana Press, New Jersey, USA.

Woster, P.M. and Casero, R.A. (2011) Polyamines Drug Discovery, Royal Chemical Society Publishing, London, UK.