283
Views
30
CrossRef citations to date
0
Altmetric
Original Research

Comparative analysis between four model nanoformulations of amphotericin B-chitosan, amphotericin B-dendrimer, betulinic acid-chitosan and betulinic acid-dendrimer for treatment of Leishmania major: real-time PCR assay plus

, , , , , & show all
Pages 7593-7607 | Published online: 24 Sep 2019

References

  • Ponte-Sucre A, Gamarro F, Dujardin J-C, et al. Drug resistance and treatment failure in leishmaniasis: a 21st century challenge. PLoS Negl Trop Dis. 2017;11(12):e0006052. doi:10.1371/journal.pntd.000605229240765
  • Alvar J, Velez ID, Bern C, et al. Leishmaniasis worldwide and global estimates of its incidence. PLoS One. 2012;7(5):e35671. doi:10.1371/journal.pone.003567122693548
  • Galvão EL, Rabello A, Cota GF. Efficacy of azole therapy for tegumentary leishmaniasis: a systematic review and meta-analysis. PLoS One. 2017;12(10):e0186117. doi:10.1371/journal.pone.018611729016694
  • Ribeiro TG, Franca JR, Fuscaldi LL, et al. An optimized nanoparticle delivery system based on chitosan and chondroitin sulfate molecules reduces the toxicity of amphotericin B and is effective in treating tegumentary leishmaniasis. Int J Nanomedicine. 2014;9:5341–5353. doi:10.2147/IJN.S6896625429219
  • Tripathi P, Jaiswal AK, Dube A, Mishra PR. Hexadecylphosphocholine (Miltefosine) stabilized chitosan modified Ampholipospheres as prototype co-delivery vehicle for enhanced killing of L. donovani. Int J Biol Macromol. 2017;105:625–637. doi:10.1016/j.ijbiomac.2017.07.07628716750
  • Yu B, Okano T, Kataoka K, Kwon G. Polymeric micelles for drug delivery: solubilization and haemolytic activity of amphotericin B. J Controlled Release. 1998;53(1):131–136.
  • Tripathi P, Dwivedi P, Khatik R, et al. Development of 4-sulfated N-acetyl galactosamine anchored chitosan nanoparticles: a dual strategy for effective management of Leishmaniasis. Colloids Surf B. 2015;136:150–159. doi:10.1016/j.colsurfb.2015.08.037
  • Cole P, Bishop J, Beckstead J, Titus R, Ryan R. Effect of amphotericin B nanodisks on Leishmania major infected mice. Pharm Anal Acta. 2014;5:1–13.
  • Meira CS, Barbosa-Filho JM, Lanfredi-Rangel A, Guimarães ET, Moreira DRM, Soares MBP. Antiparasitic evaluation of betulinic acid derivatives reveals effective and selective anti-Trypanosoma cruzi inhibitors. Exp Parasitol. 2016;166:108–115. doi:10.1016/j.exppara.2016.04.00727080160
  • Haavikko R. Synthesis of Betulin Derivatives with New Bioactivities University of Helsinki, Finland: Helsingin yliopisto; 2015:45–49.
  • Saneja A, Kumar R, Singh A, et al. Development and evaluation of long-circulating nanoparticles loaded with betulinic acid for improved anti-tumor efficacy. Int J Pharm. 2017;531(1):153–166. doi:10.1016/j.ijpharm.2017.08.07628823888
  • Alakurtti S, Heiska T, Kiriazis A, Sacerdoti-Sierra N, Jaffe CL, Yli-Kauhaluoma J. Synthesis and anti-leishmanial activity of heterocyclic betulin derivatives. Bioorg Med Chem. 2010;18(4):1573–1582. doi:10.1016/j.bmc.2010.01.00320116263
  • Domínguez-Carmona D, Escalante-Erosa F, García-Sosa K, et al. Antiprotozoal activity of betulinic acid derivatives. Phytomedicine. 2010;17(5):379–382. doi:10.1016/j.phymed.2009.08.00219748254
  • Chowdhury S, Mukherjee T, Sengupta S, Chowdhury SR, Mukhopadhyay S, Majumder HK. Novel betulin derivatives as antileishmanial agents with mode of action targeting type IB DNA topoisomerase. Mol Pharmacol. 2015;80(4):694–703. doi:10.1124/mol.111.072785
  • Chowdhury AR, Mandal S, Goswami A, et al. Dihydrobetulinic acid induces apoptosis in Leishmania donovani by targeting DNA topoisomerase I and II: implications in antileishmanial therapy. Mol Med. 2003;9(1–2):26–36.12765337
  • Pundir S, Badola A, Sharma D. Sustained release matrix technology and recent advance in matrix drug delivery system: a review. Int J Drug Res Technol. 2017;3(1):8.
  • Koohi Moftakhari Esfahani M, Alavi SE, Shahbazian S, Ebrahimi Shahmabadi H. Drug delivery of cisplatin to breast cancer by polybutylcyanoacrylate nanoparticles. Adv Polymer Technol. 2018;37(3):674–678. doi:10.1002/adv.2018.37.issue-3
  • Doun SKB, Alavi SE, Esfahani MKM, Shahmabadi HE, Alavi F, Hamzei S. Efficacy of Cisplatin-loaded poly butyl cyanoacrylate nanoparticles on the ovarian cancer: an in vitro study. Tumor Biol. 2014;35(8):7491–7497. doi:10.1007/s13277-014-1996-8
  • Shahabi J, Shahmabadi HE, Alavi SE, et al. Effect of gold nanoparticles on properties of nanoliposomal hydroxyurea: an in vitro study. Indian J Clin Biochem. 2014;29(3):315–320. doi:10.1007/s12291-013-0355-724966479
  • Dehghani F, Hoseini MHM, Memarnejadian A, et al. Immunomodulatory activities of chitin microparticles on leishmania major-infected murine macrophages. Arch Med Res. 2011;42(7):572–576. doi:10.1016/j.arcmed.2011.11.00522154683
  • Malik A, Gupta M, Mani R, Gogoi H, Bhatnagar R. Trimethyl chitosan nanoparticles encapsulated protective antigen protects the mice against anthrax. Front Immunol. 2018;9:562. doi:10.3389/fimmu.2018.0056229616046
  • Malik A, Gupta M, Gupta V, Gogoi H, Bhatnagar R. Novel application of trimethyl chitosan as an adjuvant in vaccine delivery. Int J Nanomedicine. 2018;13:7959. doi:10.2147/IJN.S17762730538470
  • Salah R. Antileishmanial activities of chitin and chitosan prepared from shrimp shell waste. Int Conf Chem Mettalurgy Environ Eng. 2015;29:177–280.
  • Jain V, Gupta A, Pawar VK, et al. Chitosan-assisted immunotherapy for intervention of experimental leishmaniasis via amphotericin B-loaded solid lipid nanoparticles. Appl Biochem Biotechnol. 2014;174(4):1309–1330. doi:10.1007/s12010-014-1084-y25106894
  • Wang JJ, Zeng ZW, Xiao RZ, et al. Recent advances of chitosan nanoparticles as drug carriers. Int J Nanomedicine. 2011;6:765. doi:10.2147/IJN.S2564621589644
  • MubarakAli D, LewisOscar F, Gopinath V, Alharbi NS, Alharbi SA, Thajuddin N. An inhibitory action of chitosan nanoparticles against pathogenic bacteria and fungi and their potential applications as biocompatible antioxidants. Microb Pathog. 2018;114:323–327. doi:10.1016/j.micpath.2017.11.04329229504
  • Singh PK, Pawar VK, Jaiswal AK, et al. Chitosan coated PluronicF127 micelles for effective delivery of amphotericin B in experimental visceral leishmaniasis. Int J Biol Macromol. 2017;105:1220–1231. doi:10.1016/j.ijbiomac.2017.07.16128780414
  • Xu L, Zhang H, Wu Y. Dendrimer advances for the central nervous system delivery of therapeutics. ACS Chem Neurosci. 2013;5(1):2–13. doi:10.1021/cn400182z24274162
  • Kalomiraki M, Thermos K, Chaniotakis NA. Dendrimers as tunable vectors of drug delivery systems and biomedical and ocular applications. Int J Nanomedicine. 2016;11:1–12. doi:10.2147/IJN.S9306926730187
  • Jain K, Verma AK, Mishra PR, Jain NK. Characterization and evaluation of amphotericin B loaded MDP conjugated poly (propylene imine) dendrimers. Nanomed. 2015;11(3):705–713. doi:10.1016/j.nano.2014.11.008
  • Hypercube Incorporation. HyperChem (TM) Professional 8.0. Gainesville, FL: Hypercube, Inc.; 2007.
  • Molecular Graphics Laboratory, Department of Molecular Biology, The Scripps Research Institute, MB-510550 N. Torrey Pines Rd. La Jolla, CA 92037-1000U.S.A. Available from: http://autodock.scripps.edu/faqs-help/tutorial/using-autodock-4-with-autodocktools.
  • Zadeh Mehrizi T, Shafiee Ardestani M, Haji Molla Hoseini M, Khamesipour A, Mosaffa N, Ramezani A. Novel nano-sized chitosan amphotericin B formulation with considerable improvement against Leishmania major. Nanomedicine (Lond). 2018;13(24):3129–3147. doi:10.2217/nnm-2018-006330463469
  • Zadeh Mehrizi T, Shafiee Ardestani M, Haji Molla Hoseini M, Khamesipour A, Mosaffa N, Ramezani A. Novel nanosized chitosan-betulinic acid against resistant Leishmania major and first clinical observation of such parasite in kidney. Sci Rep. 2018;8(1):11759. doi:10.1038/s41598-018-30103-730082741
  • Zadeh Mehrizi T, Shafiee Ardestani M, Khamesipour A, et al. Reduction toxicity of amphotericin B through loading into a novel nanoformulation of anionic linear globular dendrimer for improve treatment of leishmania major. J Mate Sci. 2018;29(8):125.
  • Zadeh Mehrizi T, Shafiee Ardestani M, Khamesipour A, et al. Reduction toxicity of Amphotericin B through loading into a novel nanoformulation of anionic linear globular dendrimer for improve treatment of leishmania major. Journal of Materials Science: Materials in Medicine 2018;29(8):125.
  • Safari J, Azizi F, Sadeghi M. Chitosan nanoparticles as a green and renewable catalyst in the synthesis of 1, 4-dihydropyridine under solvent-free conditions. N J Chem. 2015;39(3):1905–1909. doi:10.1039/C4NJ01730G
  • Jain A, Thakur K, Sharma G, Kush P, Jain UK. Fabrication, characterization and cytotoxicity studies of ionically cross-linked docetaxel loaded chitosan nanoparticles. Carbohydr Polym. 2016;137:65–74. doi:10.1016/j.carbpol.2015.10.01226686106
  • Mohammadi E, Amanlou M, Ebrahimi SES, et al. Cellular uptake, imaging and pathotoxicological studies of a novel Gd [III]–DO3A-butrol nano-formulation. RSC Adv. 2014;4(86):45984–45994. doi:10.1039/C4RA05596A
  • Ghoreishi SM, Bitarafan-Rajabi A, Azar AD, Ardestani MS, Novel AA. 99mTc-radiolabeled anionic linear globular PEG-based dendrimer-chlorambucil: non-invasive method for in-vivo biodistribution. Drug Res. 2017;67(03):149–155. doi:10.1055/s-0042-118448
  • Morakul B, Suksiriworapong J, Chomnawang MT, Langguth P, Junyaprasert VB. Dissolution enhancement and in vitro performance of clarithromycin nanocrystals produced by precipitation–lyophilization–homogenization method. Eur J Pharm Biopharm. 2014;88(3):886–896. doi:10.1016/j.ejpb.2014.08.01325201298
  • Koch S, Kessler M, Mandel K, Dembski S, Heuze K, Hackenberg S. Polycarboxylate ethers: the key towards non-toxic TiO 2 nanoparticle stabilisation in physiological solutions. Colloids Surf B. 2016;143:7–14. doi:10.1016/j.colsurfb.2016.03.010
  • Mahmoudvand H, Kheirandish F, Mirbadie SR, et al. The potential use of methotrexate in the treatment of cutaneous Leishmaniasis: in vitro assays against sensitive and meglumine antimoniate-resistant strains of Leishmania tropica. Iran J Parasitol. 2017;12(3):339.28979343
  • Abamor ES. Antileishmanial activities of caffeic acid phenethyl ester loaded PLGA nanoparticles against Leishmania infantum promastigotes and amastigotes. In Vitro. Asian Pacific J Trop Med. 2017;10(1):25–34. doi:10.1016/j.apjtm.2016.12.006
  • Vellozo NS, Pereira-Marques ST, Cabral-Piccin MP, et al. All-trans retinoic acid promotes an M1-to M2-phenotype shift and inhibits macrophage-mediated immunity to Leishmania major. Front Immunol. 2017;8:1560–1571. doi:10.3389/fimmu.2017.0156029204144
  • Marinho FA, Sangenito LS, Oliveira SS, et al. The potent cell permeable calpain inhibitor MDL28170 affects the interaction of Leishmania amazonensis with macrophages and shows anti-amastigote activity. Parasitol Int. 2017;66(5):579–583. doi:10.1016/j.parint.2017.06.01028663009
  • Davoudi N, Khamesipour A, Mahboudi F, McMaster WR. A dual drug sensitive L. major induces protection without lesion in C57BL/6 mice. PLoS Negl Trop Dis. 2014;8(5):e2785. doi:10.1371/journal.pntd.000278524873970
  • Kheirandish F, Mahmoudvand H, Khamesipour A, et al. The therapeutic effects of olive leaf extract on Leishmania major infection in BALB/c mice. Marmara Pharm J. 2017;21(4):837–842. doi:10.12991/mpj.2017.6
  • Hoseini MHM, Moradi M, Alimohammadian MH, Shahgoli VK, Darabi H, Rostami A. Immunotherapeutic effects of chitin in comparison with chitosan against Leishmania major infection. Parasitol Int. 2016;65(2):99–104. doi:10.1016/j.parint.2015.10.00726518128
  • Ghotloo S, Mollahoseini MH, Najafi A, Yeganeh F. Comparison of parasite burden using real-time polymerase chain reaction assay and limiting dilution assay in leishmania major infected mouse. Iran J Parasitol. 2015;10(4):571.26811723
  • Hajjaran H, Kazemi‐Rad E, Mohebali M, et al. Expression analysis of activated protein kinase C gene (LACK 1) in antimony sensitive and resistant Leishmania tropica clinical isolates using real‐time RT‐PCR. Int J Dermatol. 2016;55(9):1020–1026. doi:10.1111/ijd.1332127336481
  • Zadeh Mehrizi T, Mosaffa N, Haji MollaHoseini M, Shafiee Ardestani M, Khamesipour A, Ramezani A. In vivo therapeutic effects of four synthesized antileishmanial nanodrugs in the treatment of Leishmaniasis. Arch Clin Infect Dis. 2018;13(5):e80314.
  • Huynh CT, Lee DS. Controlled release. Encycl Polym Nanomate. 2015;4(Springer):439–449.
  • Tamizharasi S, Dubey A, Rathi V, Rathi J. Development and characterization of niosomal drug delivery of gliclazide. J Young Pharm. 2009;1(3):205. doi:10.4103/0975-1483.57065
  • Irby D, Du C, Li F. Lipid–drug conjugate for enhancing drug delivery. Mol Pharm. 2017;14(5):1325–1338. doi:10.1021/acs.molpharmaceut.6b0102728080053
  • Barzegar-Jalali M, Adibkia K, Valizadeh H, et al. Kinetic analysis of drug release from nanoparticles. J Pharm Pharm Sci. 2008;11(1):167–177.18471378
  • Ribeiro TG, Fumagalli MAC, Valadares DG, et al.Novel targeting using nanoparticles: an approach to the development of an effective anti-leishmanial drug-delivery system. Int J Nanomedicine, 2014;9:877–890.
  • Jain K, Verma AK, Mishra PR, Jain NK. Surface-engineered dendrimeric nanoconjugates for macrophage-targeted delivery of amphotericin B: formulation development and in vitro and in vivo evaluation. Antimicrob Agents Chemother. 2015;59(5):2479–2487. doi:10.1128/AAC.04213-1425645852
  • Kaminskas LM, Boyd BJ, Porter CJ. Dendrimer pharmacokinetics: the effect of size, structure and surface characteristics on ADME properties. Nanomedicine. 2011;6(6):1063–1084. doi:10.2217/nnm.11.6721955077
  • Gupta PK, Jaiswal AK, Asthana S, et al. Self assembled ionically sodium alginate cross-linked amphotericin B encapsulated glycol chitosan stearate nanoparticles: applicability in better chemotherapy and non-toxic delivery in visceral leishmaniasis. Pharm Res. 2015;32(5):1727–1740. doi:10.1007/s11095-014-1571-425425053
  • Dash M, Chiellini F, Ottenbrite R, Chiellini E. Chitosan—A versatile semi-synthetic polymer in biomedical applications. Prog Polym Sci. 2011;36(8):981–1014. doi:10.1016/j.progpolymsci.2011.02.001
  • Sousa MC, Varandas R, Santos RC, Santos-Rosa M, Alves V, Salvador JA. Antileishmanial activity of semisynthetic lupane triterpenoids betulin and betulinic acid derivatives: synergistic effects with miltefosine. PLoS One. 2014;9(3):e89939. doi:10.1371/journal.pone.008993924643019
  • Daftarian PM, Stone GW, Kovalski L, et al. A targeted and adjuvanted nanocarrier lowers the effective dose of liposomal amphotericin B and enhances adaptive immunity in murine cutaneous leishmaniasis. J Infect Dis. 2013;208(11):1914–1922. doi:10.1093/infdis/jit37823901083
  • Asthana S, Jaiswal AK, Gupta PK, Pawar VK, Dube A, Chourasia MK. Immunoadjuvant chemotherapy of visceral leishmaniasis in hamsters using amphotericin B-encapsulated nanoemulsion template-based chitosan nanocapsules. Antimicrob Agents Chemother. 2013;57(4):1714–1722. doi:10.1128/AAC.01984-1223357762
  • Shahnaz G, Edagwa BJ, McMillan J, et al. Development of mannose-anchored thiolated amphotericin B nanocarriers for treatment of visceral leishmaniasis. Nanomedicine. 2017;12(2):99–115. doi:10.2217/nnm-2016-032527879160