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

Tumor-targeting drug delivery of new-generation taxoids

    ,
    Edison S Zuniga

    Department of Chemistry, Stony Brook University, Stony Brook, NY 11794–3400, USA

    ,
    William T Berger

    Department of Chemistry, Stony Brook University, Stony Brook, NY 11794–3400, USA

    &
    Joshua D Seitz

    Department of Chemistry, Stony Brook University, Stony Brook, NY 11794–3400, USA

    Published Online:https://doi.org/10.4155/fmc.11.167

    A long-standing problem of conventional cancer chemotherapy is the lack of tumor specificity. Tumor-targeting drug-delivery systems have been explored to overcome this problem. These systems combine a powerful cytotoxic anticancer agent with a tumor-targeting molecule via a ‘smart’ linker to form highly efficacious drug conjugates. These drug conjugates can deliver potent cytotoxic drugs specifically to tumors and tumor cells with minimal systemic toxicity. This review describes our groups’ research on the molecular approaches to the design and development of a novel drug-delivery system bearing highly potent new-generation taxoids for tumor-targeting chemotherapy in our laboratory.

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

    References

    • Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J. Clin.61,69–90 (2011).
    • Siegel R, Ward E, Brawley O, Jemal A. Cancer statistics, 2011. CA Cancer J. Clin.61,212–236 (2011).
    • Jaracz S, Chen J, Kuznetsova LV, Ojima I. Recent advances in tumor-targeting anticancer drug conjugates. Bioorg. Med. Chem.13(17),5043–5054 (2005).
    • Ducry L, Stump B. Antibody–drug conjugates: linking cytotoxic payloads to monoclonal antibodies. Bioconjugate Chem.21,5–13 (2010).
    • Xia W, Low PS. Folate-targeted therapies for cancer. J. Med. Chem.53,6811–6824 (2010).
    • Farokhzad C, Cheng J, Teply BA et al. Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivo. Proc. Natl Acad. Sci. USA103,6315–6320 (2006).
    • Ojima I. Use of fluorine in the medicinal chemistry and chemical biology of bioactive compounds: a case study on fluorinated taxane anticancer agents. Chem. Biochem.5,628–635 (2004).
    • Ojima I. Guided molecular missiles for tumor-targeting chemotherapy: case studies using the second-generation taxoid as warheads. Acc. Chem. Res.41(1),108–119 (2008).▪ Concise review of the discovery and development of efficient mechanism-based tumor-targeting drug-delivery systems, termed ‘guided molecular missiles.’
    • Chen S, Zhao X, Chen J et al. Mechanism-based tumor-targeting drug delivery system. Validation of efficient vitamin receptor-mediated endocytosis and drug release. Bioconjugate Chem.21,979–987 (2010).▪▪ Describes a highly efficient tumor-targeting drug-delivery system for new-generation taxoids. Mechanism of internalization and drug release was verified by using fluorescent probes.
    • 10  Vredenburg MR, Ojima I, Veith J et al. Effects of orally active taxanes on P-glycoprotein modulation and colon and breast carcinoma drug resistance. J. Natl Cancer Inst.93,1234–1245 (2001).
    • 11  Ojima I, Slater JC, Michaud E et al. Syntheses and structure–activity relationships of the second-generation antitumor taxoids: exceptional activity against drug-resistant cancer cells. J. Med. Chem.39,3889–3896 (1996).
    • 12  Ojima I, Slater JC, Kuduk SD et al. Syntheses and structure–activity relationships of taxoids derived from 14-β-hydroxy-10-deacetylbaccatin III. J. Med. Chem.40(3),267–278 (1997).
    • 13  Ojima I, Wang T, Miller ML et al. Syntheses and structure-activity relationships of new second-generation taxoids. Bioorg. Med. Chem. Lett.9,3423–3428 (1999).
    • 14  Ojima I, Lin S, Wang T. The recent advances in the medicinal chemistry of taxoids with novel β-amino acid side chains. Curr. Med. Chem.6,927–954 (1999).
    • 15  Ferlini C, Distefano M, Pignatelli F et al. Antitumor activity of novel taxanes that act as cytotoxic agents and P-glycoprotein inhibitors at the same time. Brit. J. Cancer83,1762–1768 (2000).
    • 16  Geney R, Chen J, Ojima I. Recent advance in the new generation taxane anticancer agents. Med. Chem.1,125–139 (2005).
    • 17  Ehrlichova M, Vaclavikova R, Ojima I et al. Transport and cytotoxicity of paclitaxel, docetaxel and novel taxanes in human breast cancer cells. Arch. Pharmacol.372,95–105 (2005).
    • 18  Ojima I, Chen J, Sun L et al. Design, synthesis and biological evaluation of new-generation taxoids. J Med. Chem.51,3203–3221 (2008).▪▪ Reports the development of an efficient mechanism-based tumor-targeting drug delivery system, based on tumor-specific vitamin receptor-mediated endocytosis.
    • 19  Kovar J, Ehrlichova M, Smejkalova B, Zanardi I, Ojima I, Gut I. Comparison of cell death-inducing effect of novel taxane SB-T-1216 and paclitaxel in breast cancer cells. Anticancer Res.29,2951–2960 (2009).
    • 20  Beer M, Lenaz L, Amadori D. Phase II study of ortataxel in taxane-resistant breast cancer. J. Clin. Oncol. (Suppl.)26,1066 (2008).
    • 21  Kamb A, Wee S, Lengauer C. Why is cancer drug discovery so difficult?. Nat. Rev. Drug Discov.6,115–120 (2007).
    • 22  Dalerba P, Cho RW, Clarke MF. Cancer stem cells: models and concepts. Annu. Rev. Med.58,267–284 (2007).
    • 23  Botchkina GI, Zuniga ES, Das M et al. New-generation taxoid SB-T-1214 inhibits stem cell-related gene expression in 3D cancer spheriods induced by purified colon tumor-initiating cells. Mol. Cancer9,192–204 (2010).▪ Study revealed that a second-generation taxoid, SB-T-1214, effectively inhibited cancer stem cell growth and suppressed the expression of stem cell-related genes.
    • 24  Sauer LA, Dauchy RT, Blask DE. Mechanism for the antitumor and anticachectic effects of n-3 fatty acids. Cancer Res.60,5289–5295 (2000).
    • 25  Wigmore SJ, Ross JA, Falconer JS et al. The effect of polyunsaturated fatty acids on the progress of cachexia in patients with pancreatic cancer. Nutrition12,S27–S30 (1996).
    • 26  Hawkins RA, Sangster K, Arends MJ. Apoptotic death of pancreatic cancer cells induced by polyunsaturated fatty acids varies with double bond number an involves an oxidative mechanism. J. Pathol.185,61–70 (1998).
    • 27  Grammatikos SI, Subbaiah PV, Victor TA, Miller WM. n-3 and n-6 fatty acid processing and growth effects in neoplastic and noncancerous human mammary epithelial cell lines. Br. J. Cancer70,219–227 (1994).
    • 28  Diomede L, Colotta F, Piovani B, Re F, Modest EJ, Salmona M. Induction of apoptosis in human leukemic cells by the ether lipid 1-octadecyl-2-methyl-racglycero-3-phosphocholine. A possible basis for its selective action. Int. J. Cancer Res.53,124–130 (1993).
    • 29  Bradley MO, Webb NL, Anthony FH et al. Tumor targeting by covalent conjugation of a natural fatty acid to taxol. Clin. Cancer Res.7,3229–3238 (2001).▪ Presents the full preclinical evaluation of docosahexaenoic acid–paclitaxel.
    • 30  Sparreboom A, Wolff AC, Verweij J et al. Dispostition of docosahexaenoic acid: paclitaxel, a novel taxane, in blood. Clin. Cancer Res.9,151–159 (2003).
    • 31  Ibrahim NK, Desai N, Legha S et al. Phase I and pharmacokinetic study ABI-007, a cremophor-free, protein-stabilized nanoparticle formulation of paclitaxel. Clin. Cancer Res.8,1038–1044 (2002).
    • 32  Desai N, Trieu V, Yao Z et al. Increased antitumor activity, intratumor paclitaxel concentrations, and endothelial cell transport of cremophor-free, albumin-bound paclitaxel, ABI-007, compared with cremophor-based paclitaxel. Clin. Cancer Res.12,1317–1324 (2006).
    • 33  Seitz JD, Ojima I. Drug conjugates with polyunsaturated fatty acids. In: Drug Delivery in Oncology: From Basic Research to Cancer Therapy. Kratz F, Senter P, Steinhagen H (Eds). Wiley-VCH, Weinheim, Germany 1323–1360 (2011).
    • 34  Rose DP, Connolly JM. Omega-3 fatty acids as cancer chemopreventive agents. Pharmacol. Ther.83,217–244 (1999).
    • 35  Whelan J. Targeted taxane therapy for cancer. Drug Discov. Today7,90–92 (2002).
    • 36  Kuznetsova L, Chen J, Sun X et al. Syntheses and evaluation of novel fatty acid: 2nd-generation taxoid conjugates as promising anticancer agents. Bioorg. Med. Chem. Lett.16,974–977 (2006).▪▪ Reports the in vivo evaluation of polyunsaturated fatty acid–taxoid drug conjugates, which exhibited strong activity against highly multidrug-resistant colon cancer xenografts in mice.
    • 37  Chari RVJ. Targeted delivery of chemotherapeuitcs: tumor-activated prodrug therapy. Adv. Drug Deliv. Rev.31,89–104 (1998).▪ Discusses potent and tumor-specific monoclonal antibody–drug conjugates, which act as tumor-activated prodrugs for chemotherapy.
    • 38  Doronina SO, Mendelsohn BA, Bovee TD et al. Enhanced activity of monomethylauristatin F through monoclonal antibody delivery: effects of linker technology on efficacy and toxicity. Bioconjugate Chem.17,114–124 (2006).
    • 39  Beck A, Haeuw JF, Wurch T, Goetsch L, Bailly C, Corvaia N. The next generation of antibody–drug conjugates comes of age. Discov. Med.10,329–339 (2010).
    • 40  Kigawa J, Minagawa Y, Kanamori Y et al. Glutathione concentration may be a useful predictor of response to second-line chemotherapy in patients with ovarian cancer. Cancer82,697–702 (1998).
    • 41  Ojima I, Geng X, Wu X et al. Tumor-specific novel taxoid–monoclonal antibody conjugates. J Med. Chem.45,5620–5623 (2002).▪ Reports the synthesis and biological evaluation of first-generation mAb–taxoid drug conjugates, which exhibited remarkable tumor-specific anti-tumor activity in vivo.
    • 42  Chen J, Chen S, Zhao X, Kuznetsova L, Wong SS, Ojima I. Functionalized single-walled carbon nanotubes as rationally designed vehicles for tumor-targeted drug delivery. J. Am. Chem. Soc.130,16778–16785 (2008).▪▪ Reports the successful use of covalently multifunctionalized single-walled carbon nanotubes as unique vehicle for tumor-targeting drug-delivery systems. Excellent cancer cell-specificity and potency of a vitamin–single-walled carbon nanotube–taxoid conjugate was demonstrated.
    • 43  Weinstein SJ, Hartman TJ, Stolzenberg-Solomon R et al. Null association between prostate cancer and serum folate, vitamin B6, vitamin B12 and homocysteine. Cancer Epidemiol. Biomarkers Prev.12,1271–1272 (2003).
    • 44  Reddy JA, Leamon CP. Folate receptor targeted cancer chemotherapy. In: Targeted Drug Strategies for Cancer and Inflammation. Jackman AL, Leamon CP (Eds). Springer Science+Business Media, LLC, NY, USA 135–150 (2011).▪ Provides up-to-date summary of the promising preclinical evaluations of folate receptor-targeted drug conjugates.
    • 45  Leamon CP, Reddy JA. Folate-targeted chemotherapy. Adv. Drug Deliv. Rev.56,1127–1141 (2004).
    • 46  Lu Y, Low PS. Folate-mediated delivery of macromolecular anticancer therapeutic agents. Adv. Drug Deliv. Rev.54,675–693 (2002).
    • 47  Lu Y, Sega E, Leamon CP, Low PS. Folate receptor-targeted immunotherapy of cancer: mechanism and therapeutic potential. Adv. Drug Deliv. Rev.56,1161–1176 (2004).
    • 48  Leamon CP, Reddy JA, Vlahov IR et al. Synthesis and biological evaluation of EC72: a new folate-targeted chemotherapeutic. Bioconjugate Chem.16(16),803–811 (2005).
    • 49  Reddy JA, Westrick E, Vlahov I, Howard SJ, Santhapuram HK, Leamon CP. Folate receptor specific anti-tumor activity of folate–mitomycin conjugates. Cancer Chemother. Pharmacol.58,229–236 (2006).
    • 50  Zempleni J, Wijeratne SS, Hassan YI. Biotin. Biofactors35(1),36–46 (2009).
    • 51  Russell-Jones G, McTavish K, McEwan J, Rice J, Nowotnik D. Vitamin-mediated targeting as a potential mechanism to increase drug uptake by tumours. J. Inorg. Biochem.98(10),1625–1633 (2004).▪▪ Discusses the overexpression of vitamin receptors (folate, vitamin B12 and biotin) on cancer cell surfaces of a variety of cancer cell lines. Discovery of the biotin receptors as viable biomarker for tumor-targeting drug delivery.
    • 52  Mukherjee S, Ghosh RN, Masfield FR. Endocytosis. Physiol. Rev.77,759–803 (1997).
    • 53  Zuniga E, Ojima I. Design, synthesis and biological evaluation of tumor-targeting drug conjugates bearing dual warheads. Presented at: The American Chemical Society National Meeting. Denver, CO, USA, 28 August–1 September 2011.
    • 54  Prato M, Kostarelos K, Bianco A. Functionalized carbon nanotubes in drug design and discovery. Acc. Chem. Res.41(1),60–68 (2008).▪ Critical and concise review of the development of functionalized carbon nanotubes and their applications to biomedical purposes.
    • 55  Bianco A, Kostarelos K, Prato M. Applications of carbon nanotubes in drug delivery. Curr. Opin Chem. Biol.9(6),674–679 (2005).
    • 56  Kam NWS, Jessop TC, Wender PA, Dai H. Nanotube molecular transporters internalization of carbon-nanotube-protein conjugates into mammalian cells. J. Am. Chem. Soc.126,6850–6851 (2004).
    • 57  Duncan R. The dawning era of polymer therapeutics. Nat. Rev. Drug Discov.2,347–360 (2003).
    • 58  Bottini M, Cerignoli F, Dawson MI, Magrini A, Rosato N, Mustelin T. Full-length single-walled carbon nanotubes decorated with streptavidin-conjugated quantum dots as multivalent intracellular fluorescent nanoprobes. Biomacromolecules7,2259–2263 (2006).
    • 59  Kostarelos K, Lacerda L, Pastorin G et al. Cellular uptake of functionalized carbon nanotubes is independent of functional group and cell type. Nat. Nanotechnol.2(2),108–113 (2007).
    • 101  ClinicalTrialsFeeds. www.clinicaltrialsfeeds.org/clinical-trials/results/term=Drug+Taxoprexin