We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

Prostate-specific membrane antigen-targeted liposomes specifically deliver the Zn2+ chelator TPEN inducing oxidative stress in prostate cancer cells

    Christopher H Stuart

    Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA

    Department of Molecular Medicine & Translation Science, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA

    ,
    Ravi Singh

    Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA

    Comprehensive Cancer Center at Wake Forest University, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA

    ,
    Thomas L Smith

    Department of Orthopedics, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA

    ,
    Ralph D’Agostino

    Comprehensive Cancer Center at Wake Forest University, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA

    Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA

    ,
    David Caudell

    Department of Pathology & Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA

    ,
    KC Balaji

    Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA

    Comprehensive Cancer Center at Wake Forest University, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA

    Department of Urology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA

    Wake Forest Institute of Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA

    &
    William H Gmeiner

    *Author for correspondence:

    E-mail Address: bgmeiner@wakehealth.edu

    Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA

    Department of Molecular Medicine & Translation Science, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA

    Comprehensive Cancer Center at Wake Forest University, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA

    Published Online:https://doi.org/10.2217/nnm-2015-0017

    Aim: To evaluate the potential use of zinc chelation for prostate cancer therapy using a new liposomal formulation of the zinc chelator, N,N,N’,N’-tetrakis(2-pyridylmethyl)-ethylenediamine (TPEN). Materials & methods: TPEN was encapsulated in nontargeted liposomes or liposomes displaying an aptamer to target prostate cancer cells overexpression prostate-specific membrane antigen. The prostate cancer selectivity and therapeutic efficacy of liposomal (targeted and nontargeted) and free TPEN were evaluated in vitro and in tumor-bearing mice. Results & conclusion: TPEN chelates zinc and results in reactive oxygen species imbalance leading to cell death. Delivery of TPEN using aptamer-targeted liposomes results in specific delivery to targeted cells. In vivo experiments show that TPEN-loaded, aptamer-targeted liposomes reduce tumor growth in a human prostate cancer xenograft model.

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

    References

    • 1 Kirby M, Hirst C, Crawford ED. Characterising the castration-resistant prostate cancer population: a systematic review. Int. J. Clin. Pract. 65(11), 1180–1192 (2011).
    • 2 Costello LC, Franklin RB. Novel role of zinc in the regulation of prostate citrate metabolism and its implications in prostate cancer. Prostate 35(4), 285–296 (1998).
    • 3 Costello LC, Franklin RB. The clinical relevance of the metabolism of prostate cancer; zinc and tumor suppression: connecting the dots. Mol. Cancer doi:10.1186/1476-4598-5-17 (2006) (Epub ahead of print). • Zinc is identified as an important factor for advanced prostate cancer development, and a possible novel target for future treatments.
    • 4 Golovine K, Uzzo RG, Makhov P, Crispen PL, Kunkle D, Kolenko VM. Depletion of intracellular zinc increases expression of tumorigenic cytokines VEGF, IL-6 and IL-8 in prostate cancer cells via NF-kappaB-dependent pathway. Prostate 68(13), 1443–1449 (2008).
    • 5 Shah MR, Kriedt CL, Lents NH et al. Direct intra-tumoral injection of zinc-acetate halts tumor growth in a xenograft model of prostate cancer. J. Exp. Clin. Canc. Res. doi:10.1186/1756-9966-28-84 (2009) (Epub ahead of print).
    • 6 Makhov P, Golovine K, Uzzo RG et al. Zinc chelation induces rapid depletion of the X-linked inhibitor of apoptosis (XIAP) and sensitizes prostate cancer cells to TRAIL-mediated apoptosis. Cell Death Differ. 15(11), 1745–1751 (2008).
    • 7 Carraway RE, Dobner PR. Zinc pyrithione induces ERK- and PKC-dependent necrosis distinct from TPEN-induced apoptosis in prostate cancer cells. Biochim. Biophys. Acta 1823(2), 544–557 (2012).
    • 8 Gmeiner WH, Boyacioglu O, Stuart CH, Jennings-Gee J, Balaji KC. The cytotoxic and pro-apoptotic activities of the novel fluoropyrimidine F10 towards prostate cancer cells are enhanced by Zn2þ-chelation and inhibiting the serine protease Omi/HtrA2. Prostate 75(4), 360–369 (2015).• N,N,N’,N’-tetrakis(2-pyridylmethyl)-ethylenediamine is identified to induce apoptosis in a zinc-dependent manner, and can increase apoptotic activity of other drugs in advanced prostate cancer cell lines.
    • 9 Hashemi M, Ghavami S, Eshraghi M, Booy EP, Los M. Cytotoxic effects of intra and extracellular zinc chelation on human breast cancer cells. Eur. J. Pharmacol. 557(1), 9–19 (2007).
    • 10 Mendivil-Perez M, Velez-Pardo C, Jimenez-Del-Rio M. TPEN induces apoptosis independently of Zinc chelator activity in a model of acute lymphoblastic leukemia and ex vivo acute leukemia cells through oxidative stress and mitochondria caspase-3- and AIF-dependent pathways. Oxid. Med. Cell Longev. 2012, e313275 (2012).
    • 11 Khandrika L, Kumar B, Koul S, Maroni P, Koul HK. Oxidative stress in prostate cancer. Cancer Lett. 282(2), 125–136 (2009).
    • 12 Adler M, Dinterman RE, Wannemacher RW. Protection by the heavy metal chelator N,N,N’,N’-tetrakis (2-pyridylmethyl)ethylenediamine (TPEN) against the lethal action of botulinum neurotoxin A and B. Toxicon 35(7), 1089–1100 (1997).
    • 13 Safra T, Muggia F, Jeffers S et al. Pegylated liposomal doxorubicin (doxil): reduced clinical cardiotoxicity in patients reaching or exceeding cumulative doses of 500 mg/m2. Ann. Oncol. 11(8), 1029–1033 (2000).
    • 14 Awasthi VD, Goins B, Klipper R, Phillips WT. Dual radiolabeled liposomes: biodistribution studies and localization of focal sites of infection in rats. Nucl. Med. Biol. 25(2), 155–160 (1998).
    • 15 Seo JW, Mahakian LM, Kheirolomoom A et al. Liposomal Cu-64 labeling method using bifunctional chelators: poly(ethylene glycol) spacer and chelator effects. Bioconjug. Chem. 21(7), 1206–1215 (2010).
    • 16 Cao Z, Tong R, Mishra A et al. Reversible cell-specific drug delivery with aptamer-functionalized liposomes. Angew. Chem Int. Ed. Engl. 48(35), 6494–6498 (2009).
    • 17 Kang H, O’Donoghue MB, Liu H, Tan W. A liposome-based nanostructure for aptamer directed delivery. Chem. Commun. 46(2), 249–251 (2009).
    • 18 Boyacioglu O, Stuart CH, Kulik G, Gmeiner WH. Dimeric DNA aptamer complexes for high-capacity-targeted drug delivery using pH-sensitive covalent linkages. Mol. Ther. Nucleic Acids 2(7), e107 (2013).•• The prostate-specific membrane antigen-specific aptamer SZTI01 is identified and displays differential binding to cells that express the target versus those that do not.
    • 19 Foy JW, Rittenhouse K, Modi M, Patel, M. Local tolerance and systemic safety of pegaptanib sodium in the dog and rabbit. J. Ocul. Pharmacol. Ther. 23, 452–466 (2003).
    • 20 Silver DA, Pellicer I, Fair WR, Heston WD, Cordon-Cardo C. Prostate-specific membrane antigen expression in normal and malignant human tissues. Clin. Cancer Res. 3(1), 81–85 (1997).
    • 21 Wright Jr. GL, Mayer Grob B, Haley C et al. Upregulation of prostate-specific membrane antigen after androgen-deprivation therapy. Urology 48(2), 326–334 (1996).
    • 22 Tang H, Arulsamy N, Radosz M et al. Highly active copper-based catalyst for atom transfer radical polymerization. J. Am. Chem. Soc. 128(50), 16277–16285 (2006).
    • 23 Crow JP. Dichlorodihydrofluorescein and dihydrorhodamine 123 are sensitive indicators of peroxynitrite in vitro: implications for intracellular measurement of reactive nitrogen and oxygen species. Nitric Oxide 1(2), 145–157 (1997).
    • 24 Faulkner KM, Liochev SI, Fridovich I. Stable Mn(III) porphyrins mimic superoxide dismutase in vitro and substitute for it in vivo. J. Biol. Chem. 269(38), 23471–23476 (1994).
    • 25 Batinić-Haberle I, Cuzzocrea S, Rebouças JS et al. Pure MnTBAP selectively scavenges peroxynitrite over superoxide: comparison of pure and commercial MnTBAP samples to MnTE-2-PyP in two models of oxidative stress injury, an SOD-specific Escherichia coli model and carrageenan-induced pleurisy. Free Radic. Biol. Med. 46(2), 192–201 (2009).
    • 26 Costello LC, Feng P, Milon B, Tan M, Franklin RB. Role of zinc in the pathogenesis and treatment of prostate cancer: critical issues to resolve. Prostate Cancer Prostatic Dis. 7(2), 111–117 (2004).
    • 27 Chai F, Truong-Tran AQ, Evdokiou A, Young GP, Zalewski PD. Intracellular zinc depletion induces caspase activation and p21 Waf1/Cip1 cleavage in human epithelial cell lines. J. Infect. Diss. 182(s1), S85–S92 (2000).
    • 28 López JJ, Redondo PC, Salido GM, Pariente JA, Rosado JA. N,N,N′,N′-tetrakis(2-pyridylmethyl)ethylenediamine induces apoptosis through the activation of caspases-3 and -8 in human platelets. A role for endoplasmic reticulum stress. J. Thromb. Haemost. 7(6), 992–999 (2009).
    • 29 Gorrini C, Harris IS, Mak TW. Modulation of oxidative stress as an anticancer strategy. Nat. Rev. Drug Discov. 12(12), 931–947 (2013).
    • 30 Trzeciak AR, Nyaga SG, Jaruga P, Lohani A, Dizdaroglu M, Evans MK. Cellular repair of oxidatively induced DNA base lesions is defective in prostate cancer cell lines, PC-3 and DU-145. Carcinogenesis 25(8), 1359–1370 (2004).
    • 31 Fatfat M, Merhi RA, Rahal O et al. Copper chelation selectively kills colon cancer cells through redox cycling and generation of reactive oxygen species. BMC Cancer 14, 527 (2014).
    • 32 Arslan P, Di Virgilio F, Beltrame M, Tsien RY, Pozzan T. Cytosolic Ca2+ homeostasis in Ehrlich and Yoshida carcinomas. A new, membrane-permeant chelator of heavy metals reveals that these ascites tumor cell lines have normal cytosolic free Ca2+. J. Biol. Chem. 260(5), 2719–2727 (1985).
    • 33 Haga N, Fujita N, Tsuruo T. Mitochondrial aggregation precedes cytochrome c release from mitochondria during apoptosis. Oncogene 22(36), 5579–5585 (2003).
    • 34 Lin J, Zahurak M, Beer TM et al. A non-comparative randomized Phase II study of 2 doses of ATN-224, a copper/zinc superoxide dismutase inhibitor, in patients with biochemically recurrent hormone-naïve prostate cancer. Urol. Oncol. 31(5), 581–588 (2013).