Novel formulation of a methotrexate derivative with a lipid nanoemulsion

Juliana A Moura, Claudete J Valduga, Elaine R Tavares, Iara F Kretzer, Durvanei A Maria, Raul C Maranhão, Juliana A Moura, Claudete J Valduga, Elaine R Tavares, Iara F Kretzer, Durvanei A Maria, Raul C Maranhão

Abstract

Background: Lipid nanoemulsions that bind to low-density lipoprotein receptors can concentrate chemotherapeutic agents in tissues with low-density lipoprotein receptor overexpression and decrease the toxicity of the treatment. The aim of this study was to develop a new formulation using a lipophilic derivative of methotrexate, ie, didodecyl methotrexate (ddMTX), associated with a lipid nanoemulsion (ddMTX-LDE).

Methods: ddMTX was synthesized by an esterification reaction between methotrexate and dodecyl bromide. The lipid nanoemulsion was prepared by four hours of ultrasonication of a mixture of phosphatidylcholine, triolein, and cholesteryloleate. Association of ddMTX with the lipid nanoemulsion was performed by additional cosonication of ddMTX with the previously prepared lipid nanoemulsion. Formulation stability was evaluated, and cell uptake, cytotoxicity, and acute animal toxicity studies were performed.

Results: The yield of ddMTX incorporation was 98% and the particle size of LDE-ddMTX was 60 nm. After 48 hours of incubation with plasma, approximately 28% ddMTX was released from the lipid nanoemulsion. The formulation remained stable for at least 45 days at 4°C. Cytotoxicity of LDE-ddMTX against K562 and HL60 neoplastic cells was higher than for methotrexate (50% inhibitory concentration [IC(50)] 1.6 versus 18.2 mM and 0.2 versus 26 mM, respectively), and cellular uptake of LDE-ddMTX was 90-fold higher than that of methotrexate in K562 cells and 75-fold in HL60 cells. Toxicity of LDE-ddMTX, administered at escalating doses, was higher than for methotrexate (LD(50) 115 mg/kg versus 470 mg/kg; maximum tolerated dose 47 mg/kg versus 94 mg/kg) in mice. However, the hematological toxicity of LDE-ddMTX was lower than for methotrexate.

Conclusion: LDE-ddMTX was stable, and uptake of the formulation by neoplastic cells was remarkably greater than of methotrexate, which resulted in markedly greater cytotoxicity. LDE-ddMTX is thus a promising formulation to be tested in future animal models of cancer or rheumatic disease, wherein methotrexate is widely used.

Keywords: cholesterol; didodecyl methotrexate; drug delivery; methotrexate; nanoparticles.

Figures

Figure 1
Figure 1
Esterification reaction of methotrexate in the presence of cesium carbonate.
Figure 2
Figure 2
Transmission electron microscopy of (A) lipid nanoemulsion and (B) LDE-ddMTX. Abbreviations: ddMTX, didodecyl methotrexate; LDE, lipid nanoemulsion.
Figure 3
Figure 3
In vitro release profile of ddMTX from LDE in dialysis against human plasma at 37°C (mean ± standard deviation, n = 5). Abbreviations: ddMTX, didodecyl methotrexate; LDE, lipid nanoemulsion.
Figure 4
Figure 4
Cytotoxicity of (A) LDE (o), LDE-ddMTX (●), and methotrexate (■) in (B) 562 (I) and HL60 (II) cell lines. The cells were incubated with the formulations (final concentration range 0.1–2.0 mM) for 4 hours at 37°C, and survival was quantified by enzyme-linked immunosorbent assay. Results are presented the mean ± standard error of the mean of three experiments. Abbreviations: ddMTX, didodecyl methotrexate; LDE, lipid nanoemulsion.
Figure 5
Figure 5
Uptake of LDE-ddMTX (●) and methotrexate (○) 4 hours by (A) K562 and (B) HL60 cell lines. Cells were incubated with the formulations (final concentration range 0.15–2.0 mM) for 4 hours at 37°C, lysed, and the drugs were then quantified by high-performance liquid chromatography. Results are presented the mean ± standard error of the mean of three experiments. Abbreviations: ddMTX, didodecyl methotrexate; LDE, lipid nanoemulsion.
Figure 6
Figure 6
(A) Red cell count, (B) platelet count, (C) leukocyte count, and (D) leukocyte differential count in mice after 24 hours of intravenous LDE-ddMTX or methotrexate (both at 180 mg/kg dose) administration and after intravenous LDE without methotrexate. Notes: ns indicates p > 0.05; *** indicates p < 0.001. Abbreviations: ddMTX, didodecyl methotrexate; LDE, lipid nanoemulsion; LYMPH, lymphocytes; NEUTRO, neutrophyls; EOSINO, eosinophyls; MONO, monocytes; LYMPHOBL, lymphoblast.

References

    1. Estlin EJ. Continuing therapy for childhood acute lymphoblastic leukaemia: clinical and cellular pharmacology of methotrexate, 6-mercaptopurine and 6-thioguanine. Cancer Treat Rev. 2001;27:351–363.
    1. Huennekens FM. The methotrexate story: a paradigm for development of cancer chemotherapeutic agents. Adv Enzyme Regul. 1994;34:397–419.
    1. Chabner BA, Ryan DP, Paz-Ares L, Garcia-Carbonero R, Calabresi P. Antineoplastic agents. In: Hardman JG, Limbird LE, Gilman A, editors. Goodman and Gilman’s The Pharmacological Basis of Therapeutics. 10th ed. New York, NY: McGraw-Hill; 2001.
    1. Kaasgaard T, Adresen TL, Jensen SS, Holte HO, Jensen LT, Jorgensen K. Liposomes containing alkylated methotrexate analogues for phospholipase A2 mediated tumor targeted drug delivery. Chem Phys Lipids. 2009;157:94–103.
    1. Doddoli C, Chez O, Barlési F, et al. In vitro and in vivo methotrexate disposition in alveolar macrophages: Comparison of pharmacokinetic parameters of two formulations. Int J Pharm. 2005;297:180–189.
    1. Gurdag S, Khandare J, Stapels S, Metherly LH, Kannan RM. Activity of dendrimer-methotrexate conjugates on methotrexate-sensitive and -resistant cell lines. Bioconjug Chem. 2006;17:275–283.
    1. Li Y, Kwon GS. Micelle-like structures of poly(ethylene oxide)-block-poly( 2-hydroxyethyl aspartamide)-methotrexate conjugates. Colloids and Surfaces B: Biointerfaces. 1999;16:217–226.
    1. Seo DH, Jeong YI, Kim DG, Jang MJ, Jang MK, Nah JW. Methotrexate-incorporated polymeric nanoparticles of methoxypoly(ethyleneglycol)- grafted chitosan. Colloids Surf B Biointerfaces. 2009;69:157–163.
    1. Bai KB, Láng O, Orbán E, et al. Design, synthesis, and in vitro activity of novel drug delivery systems containing tuftsin derivatives and methotrexate. Bioconjug Chem. 2008;19:2260–2269.
    1. Paliwal R, Paliwal SR, Mishra N, Mehta A, Vyas SP. Engineered chylomicron mimicking carrier emulsion for lymph targeted oral delivery of methotrexate. Int J Pharm. 2009;380:181–188.
    1. Paliwal R, Rai S, Vaidya B, et al. Effect of lipid core material on characteristics of solid lipid nanoparticles designed for oral lymphatic delivery. Nanomedicine. 2009;5:184–191.
    1. Ruckmani K, Sivakumar M, Ganeshkumar PA. Methotrexate loaded solid lipid nanoparticles (SLN) for effective treatment of carcinoma. J Nanosci Nanotechnol. 2006;6:2991–2995.
    1. Ho YK, Smith RG, Brown MS, Goldstein JL. Low-density lipoprotein (LDL) receptor activity in human acute myelogenous leukemia cells. Blood. 1978;52:1099–1114.
    1. Masquelier M, Vitols S, Peterson C. Low-density lipoprotein as a carrier of antitumoral drugs: in vivo of drug-human low-density lipoproteins complexes in mice. Cancer Res. 1986;46:3842–3847.
    1. Masquelier M, Vitols S, Palsson M, Mars U, Larsson BS, Peterson CO. Low density lipoprotein as a carrier of cytostatics in cancer chemotherapy: study of stability of drug-carrier complexes in blood. J Drug Target. 2000;8:155–164.
    1. Lundberg B. Preparation of drug-low density lipoprotein complexes for delivery of antitumoral drugs via the low density lipoprotein pathway. Cancer Res. 1987;47:4105–4108.
    1. Vitols S, Angelin B, Ericsson SG, et al. Uptake of low density lipoproteins by human leukemic cells in vivo: relation to plasma lipoprotein levels and possible relevance for selective chemotherapy. Proc Natl Acad Sci U S A. 1990;87:2598–2602.
    1. Maranhão RC, Garicochea B, Silva EL, Llacer PD, Pileggi FJC, Chamone DAF. Increased plasma removal of microemulsions resembling the lipid phase of low-density lipoproteins (LDL) in patients with acute myeloid leukemia: a possible new strategy for the treatment of the disease. Braz J Med Biol Res. 1992;25:1003–1007.
    1. Maranhão RC, Garicochea B, Silva EL, et al. Plasma kinetics and biodistribution of a lipid emulsion resembling low-density lipoprotein in patients with acute leukemia. Cancer Res. 1994;54:4660–4666.
    1. Maranhão RC, César TB, Pedroso-Mariani SR, Hirata MH, Mesquita CH. Metabolic behavior in rats of a non-protein microemulsion resembling low density lipoprotein. Lipids. 1993;28:691–696.
    1. Hungria VTM, Latrilha MC, Rodrigues DG, Bydlowsky SP, Chiattone CS, Maranhao RC. Metabolism of a cholesterol-rich microemulsion (LDE) in patients with multiple myeloma and preliminary clinical study of LDE as drug vehicle for the treatment of the disease. Cancer Chemother Pharmacol. 2004;53:51–60.
    1. Rodrigues DG, Maria DA, Fernandes DC, et al. Improvement of paclitaxel therapeutic index by derivatization and association to a cholesterol-rich microemulsion: in vitro and in vivo studies. Cancer Chemother Pharmacol. 2005;55:565–576.
    1. Valduga CJ, Fernandes DC, Lo Prete AC, Azevedo CHM, Rodrigues DG, Maranhao RC. Use of a cholesterol-rich microemulsion that binds to low-density lipoprotein receptors as vehicle for etoposide. J Pharm Pharmacol. 2003;55:1615–1622.
    1. Lo Prete AC, Maria DA, Rodrigues DG, Valduga CJ, Ibanez OC, Maranhão RC. Evaluation in melanoma-bearing mice of an etoposide derivative associated to a cholesterol-rich nano-emulsion. J Pharm Pharmacol. 2006;58:801–808.
    1. Ades A, Carvalho JP, Graziani SR, et al. Uptake of a cholesterol-rich emulsion by neoplastic ovarian tissues. Gynecol Oncol. 2001;81:84–87.
    1. Graziani SR, Igreja FAF, Hegg R, et al. Uptake of a cholesterol-rich emulsion by breast cancer. Gynecol Oncol. 2002;85:493–497.
    1. Azevedo CH, Carvalho JP, Valduga CJ, Maranhão RC. Plasma kinetics and uptake by the tumor of a cholesterol-rich microemulsion (LDE) associated to etoposide oleate in patients with ovarian carcinoma. Gynecol Oncol. 2005;97:178–182.
    1. Dias ML, Carvalho JP, Rodrigues DG, Graziani SR, Maranhão RC. Pharmacokinetics and tumor uptake of a derivatized form of paclitaxel associated to a cholesterol-rich nanoemulsion (LDE) in patients with gynecologic cancers. Cancer Chemother Pharmacol. 2007;59:105–111.
    1. Pinheiro KV, Hungria VT, Ficker ES, Valduga CJ, Mesquita CH, Maranhao RC. Plasma kinetics of a cholesterol-rich microemulsion (LDE) in patients with Hodgkin’s lymphoma and a preliminary study on the toxicity of etoposide associated with LDE. Cancer Chemother Pharmacol. 2006;57:624–630.
    1. Rosowsky A, Forsch RA, Yu CS, Lazarus H, Beardsley GP. Methotrexate analogues. 21. Divergent influence of alkyl chain on the dihydrofolate reductase affinity and cytotoxicity of methotrexate monoesters. J Med Chem. 1984;27:605–609.
    1. Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assay. J Immunol Methods. 1983;65:55–63.
    1. Lowry OH, Rosebrough NJ, Farr AL, Randall R. Protein measurement with the Folin phenol reagent. J Biol Chem. 1951;193:265–275.
    1. Saito H, Kawagishi A, Tanaka M, et al. Coalescence of lipid emulsions in floating and freeze-thawing processes: examination of the coalescence transition state theory. J Colloid Interface Sci. 1999;219:129–134.
    1. Kabalnov A, Tarara T, Arlauskas R, Werrs J. Phospholipids as emulsion stabilizers. 2. Phase behavior versus emulsion stability. J Colloid Interface Sci. 1996;184:227–235.
    1. Pignatello R, Spampinato G, Sorrenti V, et al. Lipophilic methotrexate conjugates with antitumor activity. Eur J Pharm Sci. 2000;10:237–245.
    1. Maranhão RC, Tavares ER, Padoveze AF, Valduga CJ, Rodrigues DG, Pereira MD. Paclitaxel associated with cholesterol-rich nanoemulsions promotes atherosclerosis regression in the rabbit. Atherosclerosis. 2008;197:959–966.
    1. Ofner CM, 3rd, Pica K, Bowman BJ, Chen CS. Growth inhibition, drug load, and degradation studies of gelatin/methotrexate conjugates. Int J Pharm. 2006;308:90–99.

Source: PubMed

3
Prenumerera