Paclitaxel-loaded biodegradable ROS-sensitive nanoparticles for cancer therapy

. 2019 ; 14 () : 6269-6285. [epub] 20190806

Jazyk angličtina Země Nový Zéland Médium electronic-ecollection

Typ dokumentu časopisecké články

Perzistentní odkaz   https://www.medvik.cz/link/pmid31496685

BACKGROUND: Reactive oxygen species (ROS), such as hydrogen peroxide and superoxide, trigger biodegradation of polymer-based nanoparticles (NPs) bearing pinacol-type boronic ester groups. These NPs may selectively release their cargo, in this case paclitaxel (PTX), at the high levels of ROS present in the intracellular environment of inflamed tissues and most tumors. PURPOSE: The main objective was to determine anti-tumor efficacy of PTX-loaded ROS-sensitive NPs and to examine whether macrophage infiltration had any impact on treatment efficacy. METHODS: NPs were synthesized and their characteristics in the presence of H2O2 were demonstrated. Both confocal microscopy as well as flow cytometry approaches were used to determine degradation of ROS-sensitive NPs. HeLa cells were cultured in vitro and used to establish tumor xenografts in nude mice. In vivo experiments were performed to understand toxicity, biodistribution and anti-tumor efficacy of the NPs. Moreover, we performed immunohistochemistry on tumor sections to study infiltration of M1 and M2 subsets of macrophages. RESULTS: We demonstrated that PTX delivered in NPs containing a ROS-sensitive polymer exhibits a better anti-tumor efficacy than PTX in NPs containing ROS-non-sensitive polymer, free PTX or Abraxane® (nab-PTX). The biodistribution revealed that ROS-sensitive NPs exhibit retention in liver, spleen and lungs, suggesting a potential to target cancer metastasizing to these organs. Finally, we demonstrated a correlation between infiltrated macrophage subsets and treatment efficacy, possibly contributing to the efficient anti-tumor effects. CONCLUSION: Treatment with ROS-sensitive NPs containing PTX gave an improved therapeutic effect in HeLa xenografts than their counterpart, free PTX or nab-PTX. Our data revealed a correlation between macrophage infiltration and efficiency of the different antitumor treatments, as the most effective NPs resulted in the highest infiltration of the anti-tumorigenic M1 macrophages.

Zobrazit více v PubMed

D’Autreaux B, Toledano MB. ROS as signalling molecules: mechanisms that generate specificity in ROS homeostasis. Nat Rev Mol Cell Bio. 2007;8(10):813–824. doi:10.1038/nrm2256 PubMed DOI

Sies H, Stahl W, Sundquist AR. Antioxidant functions of vitamins - Vitamin-E and Vitamin-C, beta-carotene, and other carotenoids. Ann Ny Acad Sci. 1992;669:7–20. doi:10.1111/j.1749-6632.1992.tb17085.x PubMed DOI

Sies H. Oxidative stress - from basic research to clinical-application. Am J Med. 1991;91:S31–S38. doi:10.1016/0002-9343(91)90281-2 PubMed DOI

Minelli A, Bellezza I, Conte C, Culig Z. Oxidative stress-related aging: a role for prostate cancer? Biochim Biophys Acta. 2009;1795(2):83–91. PubMed

Giorgio M, Trinei M, Migliaccio E, Pelicci PG. Hydrogen peroxide: a metabolic by-product or a common mediator of ageing signals? Nat Rev Mol Cell Bio. 2007;8(9):722a–728a. PubMed

Di Virgilio F. New pathways for reactive oxygen species generation in inflammation and potential novel pharmacological targets. Curr Pharm Design. 2004;10(14):1647–1652. doi:10.2174/1381612043384727 PubMed DOI

Dugan LL, Quick KL. Reactive oxygen species and aging: evolving questions. Sci Aging Knowledge Environ. 2005;2005(26):pe20. doi:10.1126/sageke.2005.26.pe20 PubMed DOI

Muhammad S, Bierhaus A, Schwaninger M. Reactive oxygen species in diabetes-induced vascular damage, stroke, and alzheimer’s disease. J Alzheimers Dis. 2009;16(4):775–785. doi:10.3233/JAD-2009-0982 PubMed DOI

Touyz RM. Reactive oxygen species and angiotensin II signaling in vascular cells - implications in cardiovascular disease. Braz J Med Biol Res. 2004;37(8):1263–1273. doi:10.1590/S0100-879X2004000800018 PubMed DOI

Furukawa S, Fujita T, Shimabukuro M, et al. Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest. 2004;114(12):1752–1761. doi:10.1172/JCI21625 PubMed DOI PMC

Johar R, Sharma R, Kaur A, Mukherjee TK. Role of reactive oxygen species in estrogen dependant breast cancer complication. Anti-Cancer Agent Me. 2016;16(2):190–199. doi:10.2174/1871520615666150518092315 PubMed DOI

Sharma A, Rajappa M, Satyam A, Sharma M. Oxidant/anti-oxidant dynamics in patients with advanced cervical cancer: correlation with treatment response. Mol Cell Biochem. 2010;341(1–2):65–72. doi:10.1007/s11010-010-0437-2 PubMed DOI

Chan DW, Liu VWS, Tsao GSW, et al. Loss of MKP3 mediated by oxidative stress enhances tumorigenicity and chemoresistance of ovarian cancer cells. Carcinogenesis. 2008;29(9):1742–1750. doi:10.1093/carcin/bgn167 PubMed DOI

Edderkaoui M, Hong P, Vaquero EC, et al. Extracellular matrix stimulates reactive oxygen species production and increases pancreatic cancer cell survival through 5-lipoxygenase and NADPH oxidase. Am J Physiol Gastrointest Liver Physiol. 2005;289(6):G1137–1147. doi:10.1152/ajpgi.00508.2004 PubMed DOI

Azad N, Rojanasakul Y, Vallyathan V. Inflammation and lung cancer: roles of reactive oxygen/nitrogen species. J Toxicol Environ Health B Crit Rev. 2008;11(1):1–15. doi:10.1080/10937400701436460 PubMed DOI

Wang Z, Li Z, Ye Y, Xie L, Li W. Oxidative stress and liver cancer: etiology and therapeutic targets. Oxid Med Cell Longev. 2016;2016:7891574. doi:10.1155/2016/7891574 PubMed DOI PMC

Bur H, Haapasaari K-M, Turpeenniemi-Hujanen T, et al. Oxidative stress markers and mitochondrial antioxidant enzyme expression are increased in aggressive Hodgkin lymphomas. Histopathology. 2014;65(3):319–327. doi:10.1111/his.12389 PubMed DOI

Salganik RI, Albright CD, Rodgers J, et al. Dietary antioxidant depletion: enhancement of tumor apoptosis and inhibition of brain tumor growth in transgenic mice. Carcinogenesis. 2000;21(5):909–914. doi:10.1093/carcin/21.4.701 PubMed DOI

Klaunig JE, Xu Y, Isenberg JS, et al. The role of oxidative stress in chemical carcinogenesis. Environ Health Perspect. 1998;106(Suppl 1):289–295. PubMed PMC

Petros RA, DeSimone JM. Strategies in the design of nanoparticles for therapeutic applications. Nat Rev Drug Discov. 2010;9(8):615–627. doi:10.1038/nrd2591 PubMed DOI

Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol. 2007;2(12):751–760. doi:10.1038/nnano.2007.387 PubMed DOI

Couvreur P. Nanoparticles in drug delivery: past, present and future. Adv Drug Deliv Rev. 2013;65(1):21–23. doi:10.1016/j.addr.2012.04.010 PubMed DOI

Skotland T, Iversen TG, Sandvig K. Development of nanoparticles for clinical use. Nanomedicine (Lond). 2014;9(9):1295–1299. doi:10.2217/nnm.14.81 PubMed DOI

Pu HL, Chiang WL, Maiti B, et al. Nanoparticles with dual responses to oxidative stress and reduced ph for drug release and anti-inflammatory applications. ACS Nano. 2014;8(2):1213–1221. doi:10.1021/nn4058787 PubMed DOI

Dickinson BC, Chang CJ. A targetable fluorescent probe for imaging hydrogen peroxide in the mitochondria of living cells. J Am Chem Soc. 2008;130(30):9638–9639. doi:10.1021/ja802355u PubMed DOI PMC

Savina A, Peres A, Cebrian I, et al. The small GTPase Rac2 controls phagosomal alkalinization and antigen crosspresentation selectively in CD8(+) dendritic cells. Immunity. 2009;30(4):544–555. doi:10.1016/j.immuni.2009.01.013 PubMed DOI

de Gracia Lux C, Joshi-Barr S, Nguyen T, et al. Biocompatible polymeric nanoparticles degrade and release cargo in response to biologically relevant levels of hydrogen peroxide. J Am Chem Soc. 2012;134(38):15758–15764. doi:10.1021/ja303372u PubMed DOI PMC

Shim MS, Xia Y. A reactive oxygen species (ROS)-responsive polymer for safe, efficient, and targeted gene delivery in cancer cells. Angew Chem Int Ed Engl. 2013;52(27):6926–6929. doi:10.1002/anie.201209633 PubMed DOI PMC

Jager E, Hocherl A, Janouskova O, et al. Fluorescent boronate-based polymer nanoparticles with reactive oxygen species (ROS)-triggered cargo release for drug-delivery applications. Nanoscale. 2016;8(13):6958–6963. doi:10.1039/C6NR00791K PubMed DOI

Liou GY, Storz P. Reactive oxygen species in cancer. Free Radic Res. 2010;44(5):479–496. doi:10.3109/10715761003667554 PubMed DOI PMC

Nathan C, Cunningham-Bussel A. Beyond oxidative stress: an immunologist’s guide to reactive oxygen species. Nat Rev Immunol. 2013;13(5):349–361. doi:10.1038/nri3423 PubMed DOI PMC

Guillen J. FELASA guidelines and recommendations. J Am Assoc Lab Anim Sci. 2012;51(3):311–321. PubMed PMC

Knapp W, Dorken B, Rieber P, Schmidt RE, Stein H, von dem Borne AE. CD antigens 1989. Int J Cancer. 1989;44(1):190–191. doi:10.1002/ijc.2910440135 PubMed DOI

Khazen W, M’Bika JP, Tomkiewicz C, et al. Expression of macrophage-selective markers in human and rodent adipocytes. FEBS Lett. 2005;579(25):5631–5634. doi:10.1016/j.febslet.2005.09.032 PubMed DOI

Komohara Y, Fujiwara Y, Ohnishi K, Takeya M. Tumor-associated macrophages: potential therapeutic targets for anti-cancer therapy. Adv Drug Deliv Rev. 2016;99(Pt B):180–185. doi:10.1016/j.addr.2015.11.009 PubMed DOI

Wang C, Wang Y, Wang Y, Fan M, Luo F, Qian Z. Characterization, pharmacokinetics and disposition of novel nanoscale preparations of paclitaxel. Int J Pharm. 2011;414(1–2):251–259. doi:10.1016/j.ijpharm.2011.05.014 PubMed DOI

Storz P. Reactive oxygen species in tumor progression. Front Biosci. 2005;10:1881–1896. PubMed

Huang D, Zhuang Y, Shen H, Yang F, Wang X, Wu D. Acetal-linked PEGylated paclitaxel prodrugs forming free-paclitaxel-loaded pH-responsive micelles with high drug loading capacity and improved drug delivery. Mater Sci Eng C Mater Biol Appl. 2018;82:60–68. doi:10.1016/j.msec.2017.08.063 PubMed DOI

Lu J, Chuan X, Zhang H, et al. Free paclitaxel loaded PEGylated-paclitaxel nanoparticles: preparation and comparison with other paclitaxel systems in vitro and in vivo. Int J Pharm. 2014;471(1–2):525–535. doi:10.1016/j.ijpharm.2014.05.032 PubMed DOI

Jager E, Giacomelli FC. Soft matter assemblies as nanomedicine platforms for cancer chemotherapy: a journey from market products towards novel approaches. Curr Top Med Chem. 2015;15(4):328–344. doi:10.2174/1568026615666150130152300 PubMed DOI

Bargagli E, Olivieri C, Bennett D, Prasse A, Muller-Quernheim J, Rottoli P. Oxidative stress in the pathogenesis of diffuse lung diseases: a review. Respir Med. 2009;103(9):1245–1256. doi:10.1016/j.rmed.2009.04.014 PubMed DOI

Corzo CA, Cotter MJ, Cheng P, et al. Mechanism regulating reactive oxygen species in tumor-induced myeloid-derived suppressor cells. J Immunol. 2009;182(9):5693–5701. doi:10.4049/jimmunol.0802775 PubMed DOI PMC

Barja G. Mitochondrial oxygen consumption and reactive oxygen species production are independently modulated: implications for aging studies. Rejuvenation Res. 2007;10(2):215–224. doi:10.1089/rej.2006.0516 PubMed DOI

Logan A, Shabalina IG, Prime TA, et al. In vivo levels of mitochondrial hydrogen peroxide increase with age in mtDNA mutator mice. Aging Cell. 2014;13(4):765–768. doi:10.1111/acel.12255 PubMed DOI PMC

Beckman KB, Ames BN. The free radical theory of aging matures. Physiol Rev. 1998;78(2):547–581. doi:10.1152/physrev.1998.78.2.547 PubMed DOI

Brand MD. Uncoupling to survive? The role of mitochondrial inefficiency in ageing. Exp Gerontol. 2000;35(6–7):811–820. doi:10.1016/S0531-5565(00)00135-2 PubMed DOI

Pavelescu LA. On reactive oxygen species measurement in living systems. J Med Life. 2015;8(Spec Issue):38–42. PubMed PMC

Larsen SK, Gao Y, Basse PH. NK cells in the tumor microenvironment. Crit Rev Oncog. 2014;19(1–2):91–105. doi:10.1615/CritRevOncog.v19.i1-2 PubMed DOI PMC

Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Cell. 2010;141(1):39–51. doi:10.1016/j.cell.2010.03.014 PubMed DOI PMC

Mantovani A, Sica A, Locati M. Macrophage polarization comes of age. Immunity. 2005;23(4):344–346. doi:10.1016/j.immuni.2005.10.001 PubMed DOI

Komohara Y, Jinushi M, Takeya M. Clinical significance of macrophage heterogeneity in human malignant tumors. Cancer Sci. 2014;105(1):1–8. doi:10.1111/cas.12314 PubMed DOI PMC

Brown ER, Charles KA, Hoare SA, et al. A clinical study assessing the tolerability and biological effects of infliximab, a TNF-alpha inhibitor, in patients with advanced cancer. Ann Oncol. 2008;19(7):1340–1346. doi:10.1093/annonc/mdn054 PubMed DOI

Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140(6):883–899. doi:10.1016/j.cell.2010.01.025 PubMed DOI PMC

Lewis CE, Pollard JW. Distinct role of macrophages in different tumor microenvironments. Cancer Res. 2006;66(2):605–612. doi:10.1158/0008-5472.CAN-05-4005 PubMed DOI

Kodumudi KN, Woan K, Gilvary DL, Sahakian E, Wei S, Djeu JY. A novel chemoimmunomodulating property of docetaxel: suppression of myeloid-derived suppressor cells in tumor bearers. Clin Cancer Res. 2010;16(18):4583–4594. doi:10.1158/1078-0432.CCR-10-0613 PubMed DOI PMC

Fusser M, Overbye A, Pandya AD, et al. Cabazitaxel-loaded Poly(2-ethylbutyl cyanoacrylate) nanoparticles improve treatment efficacy in a patient derived breast cancer xenograft. J Control Release. 2018;293:183–192. doi:10.1016/j.jconrel.2018.11.027 PubMed DOI

Wanderley CW, Colon DF, Luiz JPM, et al. Paclitaxel reduces tumor growth by reprogramming tumor-associated macrophages to an M1- profile in a TLR4-dependent manner. Cancer Res. 2018. doi:10.1158/0008-5472.CAN-17-3480 PubMed DOI

Najít záznam

Citační ukazatele

Nahrávání dat ...

Možnosti archivace

Nahrávání dat ...